facultad de ciencias - core

199
Facultad de Ciencias COMPLEMENT ACTIVATION IN NON-SMALL CELL LUNG CANCER AND ITS EFFECT ON TUMOR PROGRESSION Leticia Corrales Pecino

Upload: others

Post on 16-Oct-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Facultad de Ciencias - CORE

Facu l tad de C ienc ias

C O M P L E M E N T A C T I V A T I O N I N

N O N - S M A L L C E L L L U N G C A N C E R A N D

I T S E F F E C T O N T U M O R P R O G R E S S I O N

L e t i c i a C o r r a l e s P e c i n o

Page 2: Facultad de Ciencias - CORE

Servicio de Publicaciones de la Universidad de Navarra

ISBN 84-8081-130-7

Page 3: Facultad de Ciencias - CORE
Page 4: Facultad de Ciencias - CORE

F a c u l t a d d e C i e n c i a s

Complement activation in non-small cell lung

cancer and its effect on tumor progression

Memoria presentada por Dª Leticia Corrales Pecino para aspirar al grado

de Doctor por la Universidad de Navarra

Leticia Corrales Pecino

Page 5: Facultad de Ciencias - CORE
Page 6: Facultad de Ciencias - CORE

El presente trabajo ha sido realizado bajo mi dirección en el Departamento

de Bioquímica y el Área de Oncología del CIMA y autorizo su presentación

ante el Tribunal que lo ha de juzgar.

Pamplona, 22 de febrero de 2011

Dr. Rubén Pío Osés

Page 7: Facultad de Ciencias - CORE
Page 8: Facultad de Ciencias - CORE

El presente trabajo ha sido presentado en la Facultad de Ciencias

para optar al título de Doctor por la Universidad de Navarra

Page 9: Facultad de Ciencias - CORE
Page 10: Facultad de Ciencias - CORE

Este trabajo ha sido realizado con las siguientes ayudas:

“Ayuda para la formación de personal investigador”, de la Fundación para

la Investigación Médica Aplicada.

Beca para la “Formación de Profesorado Universitario” del Ministerio de

Ciencia e Innovación.

UTE-project CIMA.

Plan Nacional de Investigación Científica, Desarrollo e Innovación

Tecnológica del Ministerio de Educación y Ciencia (SAF-2005-01302).

Programa de Promoción de la Investigación Biomédica y en Ciencias de la

Salud del Ministerio de Sanidad y Consumo (PI080923).

Page 11: Facultad de Ciencias - CORE
Page 12: Facultad de Ciencias - CORE

A mis padres

A Edu y Montse

A mi tío Jose María

Page 13: Facultad de Ciencias - CORE
Page 14: Facultad de Ciencias - CORE

Acknowledgements

Gracias en primer lugar a la Universidad de Navarra, al departamento de

Bioquímica y al área de Oncología del CIMA, por haberme dado la oportunidad de

realizar la tesis doctoral.

En segundo lugar, gracias a mi director de tesis Rubén Pío, con el que he

empezado a dar los primeros pasos en el mundo de la investigación. Gracias por todo el

tiempo dedicado y la ayuda proporcionada para que saliese adelante este trabajo.

Muchas gracias por la dirección de la tesis, por tu paciencia y por la confianza

depositada en mí.

A Luis Montuenga tengo que agradecer todos sus consejos y opiniones que han

sido de gran ayuda para realizar esta tesis.

También me gustaría dar las gracias a Daniel Ajona, una fuente inagotable de

ideas, algunas han sido indispensables en este trabajo. Gracias por tu ayuda, tus ánimos

y por tus comentarios constructivos y por supuesto, siempre “razonables”.

A todos mis compañeros del grupo de Biomarcadores con los que he pasado

muchas horas y muchos buenos momentos: Maribel, Alberto, Marta, María José, Jackie,

Ana, Elena y Uxúa. A Txiki, Cristina y Amaya quiero darles las gracias especialmente

por todo lo que pacientemente me han enseñado y ayudado. Y a los que ya se fueron:

Javi, David, Evan, Patricia, Rebeca, Iñaki y Teresa. Muchísimas gracias a todos.

No puedo olvidarme del laboratorio de la Dra. Anna Blom en Malmö. Gracias por

haberme dado la oportunidad de trabajar y aprender tanto con vosotros. Gracias también

a Marcin Okroj por toda su ayuda y apoyo durante mi estancia.

Al laboratorio de Microambiente Tumoral: Saray, Álvaro, Lili, Rafa, Xabi y los

que ya se fueron, Eva, Marta y Erik. Gracias por dejarme ser una “okupa” y luego

ascenderme a grado de hija adoptiva. Y muchas gracias Ana por tu ayuda, por darme

buenas ideas y colaborar tan activamente en este trabajo.

Quiero también agradecer a todas las personas de la planta de Oncología, a

nuestros vecinos y compañeros de cultivos 1B con los que tantas horas he pasado: y a

todos los que he ido conociendo en estos años: Alice, Leyre, Celia, Diego, Iker,

Carolina, Manu, María, Javi, Angela, Lola… ¡es imposible nombraros a todos!, vuestra

ayuda y consejos han contribuido a la realización de este trabajo. También quisiera

agradecer la ayuda que me ofrecieron otras personas del CIMA, en especial los

laboratorios del Dr. Riezu, Dr. Lasarte y Dr. Páramo.

Al Dr. Alfredo Martínez, muchas gracias por toda la ayuda y consejos que nos

ofreciste para el estudio de la angiogénesis.

Gracias a las secretarias de la planta de Oncología, Marisol y Leticia, por vuestro

trabajo que tanto facilita el nuestro.

Page 15: Facultad de Ciencias - CORE

Acknowledgements

Gracias a todas mis compañeras de piso en estos 4 años, por aguantarme y

compartir conmigo tantos buenos momentos. A Belén y Mariona, por vuestra amistad

desde que llegamos a Pamplona; a Erika, por tu alegría, generosidad y por comprender,

compartir e incluso intercambiar “frikadas”; a Gloria, Silvia y Almu, por hacerme sentir

como “en familia”; a Carmela, porque contigo el debate durante las comidas siempre

estaba asegurado; a Laura, por tu sentido del humor, tus sevillanas maneras y por tu

espíritu crítico, gracias por enseñarme a ver las cosas desde otro punto de vista; a Elena

por demostrarme que es una persona con la que siempre podré contar; y por último,

gracias a la “UNESCO”, a Mónica por tu disponibilidad y tus artes culinarias, a Susana

por tu generosidad y contagiarnos un poco tu “happy world”, y a Lili por ser tan buena

compañera de piso, de laboratorio, de planes… y tan buena amiga. No puedo olvidarme

de vuestros respectivos novios (algunos ya maridos), gracias por compartir con nosotras

muchas risas, bastantes comidas y alguna que otra excursión. En definitiva, gracias a

todos por ser mi familia en Pamplona y haber conseguido que no me encontrase sola en

ningún momento.

A Juana Mari, porque aunque fueron pocos meses de estancia en Pamplona

bastaron para empezar una amistad que estoy segura perdurará a pesar de la distancia.

Gracias por tu buen humor, tus consejos, por tus buenos detalles y en definitiva por ser

como eres, una “maravilla de bé”.

Gracias a Miriam por tu amistad, tus consejos y por decir las cosas sin rodeos, y

por ser tan sumamente servicial.

Gracias a Rudi y a Manolo por ser mucho más que mis profesores de pintura. Por

vuestra paciencia, vuestro sentido del humor, por dar siempre ánimos incluso en

momentos no muy buenos y por ser tan excelentes artistas.

Gracias a todos mis amigos de Algeciras, por vuestro apoyo y ánimos, gracias por

hacer que desconectara. Tampoco puedo olvidarme de Chico, el perro de Rubio, gracias

porque mientras viva la comunidad científica puede estar segura de que existe un animal

más feo que un ratón nude.

Gracias especialmente a Elena Domínguez, por tu optimismo y tus ánimos.

Gracias porque es tan fácil confiar en ti.

Finalmente, gracias a mi familia. A Edu y Montse, por vuestro apoyo, cariño y por

cuidar tanto y tan bien de mí. Y aunque es muy difícil resumir todo lo que os tengo que

agradecer en unas líneas, gracias papá y mamá por vuestra confianza en mí, por

escuchar los cientos de “ohu, que hoy no me ha salido el experimento” y aguantar mi

humor a veces algo agrio, por la ilusión que ponéis en todo lo que hago y por ser mi

referente a seguir. Gracias de corazón.

Page 16: Facultad de Ciencias - CORE

ABBREVIATIONS

Page 17: Facultad de Ciencias - CORE
Page 18: Facultad de Ciencias - CORE

Abbreviations

7AAD 7-amino-actinomycin D

ADCC Antibody-dependent cellular cytotoxicity

AMD Age-related macular degeneration

APC Antigen presenting cells

APH50 Alternative pathway hemolysis (50%)

ARG1 Arginase 1

ATCC American Type Culture Collection

BCR B cell antigen receptor

BEAS Bronchial epithelial cells

bFGF Basic fibroblast growth factor

BSA Bovine serum albumin

C1 INH C1 inhibitor

C3aR C3a receptor

C4BP C4-binding protein

C5aR-1 (CD88) C5a receptor-1

C5L2 C5a-like receptor 2

CCL Chemokine ligand

CCP Complement control protein

CDC Complement dependent cytotoxicity

Cdk4 Cyclin-dependent kinase 4

CFHR Complement factor H-related protein

CK2 Casein kinase 2

CR1 (CD35) Complement receptor type 1

CTLA4 (CD152) Cytotoxic T-lymphocyte antigen 4

CVF Cobra venom factor

DAF (CD55) Decay accelerating factor

DEPC Diethyl pyrocarbonate

DMSO Dimethylsulphoxide

dNTP Deoxyribonucleoside triphosphate

DTT Dithiothreitol

ECM Extracellular matrix

EDTA Ethylenediaminetetraacetic acid

EGTA Ethylene glycol tetraacetic acid

ELISA Enzyme-linked immunosorbent assay

Page 19: Facultad de Ciencias - CORE

Abbreviations

ERK Extracellular signal-regulated kinase

FACS Fluorescence activated cell sorting

FBS Fetal bovine serum

FH Factor H

FHL-1 Factor H-like protein 1

FITC Fluorescein isothiocyanate

Foxp3 Transcription factor forkhead box P3

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

GPCR G-protein-coupled receptor

GPI Glycosylphosphatidylinositol

GPI-PC Glycosylphosphatidylinositol-phospholipase C

GVB Veronal buffer saline with gelatin

HBEC Human bronchial epithelial cells

HI-NHS Heat inactivated normal human serum

HI-NMS Heat inactivated normal mouse serum

HPLC High-pressure liquid chromatography

hTERT Human telomerase reverse transcriptase

HUVEC Human umbilical vein endothelial cells

IDO Indoleamine 2,3-dioxygenase

IL Interleukin

INF-γ Interferon-gamma

iNOS Inducible nitric oxide synthase

KO Knockout

LAG3 (CD223) Lymphocyte activation gene 3

LDH Lactate dehydrogenase

LLC or 3LL Lewis lung carcinoma cells

mAb Monoclonal antibody

MAC Membrane attack complex

MAPK Mitogen-activated protein kinase

MASP MBL-associated serine protease

MBL Mannose-binding lectin

MCP (CD46) Membrane cofactor protein

MCP1 Monocyte chemotactic protein-1

mCRP Membrane complement regulatory protein

Page 20: Facultad de Ciencias - CORE

Abbreviations

MDSC Myeloid-derived suppressor cells

MFI Mean fluorescence intensity

MMP Matrix metalloproteinase

NHS Normal human serum

NK-κB Nuclear factor kappa-light-chain-enhancer of

activated B cells

NMS Normal mouse serum

NO Nitric oxide

NSCLC Non-small cell lung cancer

ONOO Peroxynitrites

PBS Phosphate buffered saline

PBST Phosphate buffered saline-Tween

PCR Polymerase chain reaction

PD1 Programmed death 1

PDL1 (B7-H1) Programmed death ligand 1

PDT Photodynamic therapy

PE Phycoerythrin

PI Phosphatidylinositol

PI-PLC Phosphatidylinositol-specific phospholipase C

PKC Protein kinase C

RNS Reactive nitrogen species

ROS Reactive oxygen species

SCLC Small cell lung cancer

SCR Short consensus repeat

SDS Sodium dodecyl sulfate

SDS-PAGE SDS polyacrylamide gel electrophoresis

TAE Tris-acetate-EDTA buffer

TAM Tumor-associated macrophage

TBS Tris buffered saline

TGF-β Transforming growth factor beta

TIMP Tissue inhibitor of metalloproteinases

TNFSF4 (OX40L) Tumor necrosis factor (ligand) superfamily

member 4

Page 21: Facultad de Ciencias - CORE

Abbreviations

TNF-α Tumor necrosis factor-alpha

Tregs Regulatory T cells

VB Veronal buffer saline

VEGF Vascular endothelial growth factor

WT Wild-type

Page 22: Facultad de Ciencias - CORE

INDEX

Page 23: Facultad de Ciencias - CORE
Page 24: Facultad de Ciencias - CORE

Index

INTRODUCTION ________________________________________________ 1

1. LUNG CANCER _______________________________________________ 3

1.1. Lung cancer classification _____________________________________________4

1.2. Lung cancer staging __________________________________________________4

1.3. Lung cancer etiology _________________________________________________6

2. THE COMPLEMENT SYSTEM __________________________________ 6

2.1. Complement activation _______________________________________________8

2.1.1 The classical pathway ______________________________________________8

2.1.2 The lectin pathway ________________________________________________9

2.1.3 The alternative pathway ____________________________________________9

2.1.4 Alternative routes of complement activation ____________________________9

2.2. Complement regulation ______________________________________________10

2.2.1 Soluble regulators of the complement system___________________________11

a. C1 inhibitor _____________________________________________________11

b. C4-binding protein _______________________________________________12

c. Factor H and factor H-like protein 1 __________________________________12

d. Vitronectin______________________________________________________13

e. Clusterin _______________________________________________________13

2.2.2 Membrane complement regulatory proteins ____________________________14

a. Complement receptor type 1 ________________________________________14

b. Membrane cofactor protein _________________________________________14

c. Decay accelerating factor __________________________________________14

d. CD59 __________________________________________________________15

2.3. Biological effects of complement activation ______________________________15

2.3.1 Cell lysis by the membrane attack complex ____________________________15

2.3.2 Opsonization of the cell by C3b and its related fragments _________________15

2.3.3 Biological effects mediated by anaphylatoxins__________________________16

3. COMPLEMENT AND CANCER ________________________________ 18

3.1. Complement in the immune surveillance against tumors_____________________18

3.2. Complement regulation on cancer cells __________________________________19

3.3. Complement in the immunotherapy of tumors_____________________________21

3.4. Complement, inflammation and cancer __________________________________21

3.5. Complement and lung cancer__________________________________________23

HYPOTHESIS AND OBJECTIVES_________________________________ 27

MATERIALS AND METHODS ____________________________________ 29

1. MATERIALS _________________________________________________ 31

1.1. Animals __________________________________________________________31

Page 25: Facultad de Ciencias - CORE

Index

1.2. Clinical samples from lung cancer patients _______________________________31

1.3. Normal human serum________________________________________________31

1.4. Plasma ___________________________________________________________32

1.5. Human non-small cell lung cancer cells__________________________________32

1.6. Immortalized normal human epithelial cells ______________________________32

1.7. Endothelial cells____________________________________________________33

1.8. Murine lung cancer cells _____________________________________________33

1.9. Hybridoma ________________________________________________________33

1.10. Factor H________________________________________________________33

1.11. C5aR-1 antagonist ________________________________________________33

2. METHODS ___________________________________________________ 34

2.1. Methods for the generation of experimental material _______________________34

2.1.1 Cell culture _____________________________________________________34

a. Cell culture conditions_____________________________________________34

b. Subcultures _____________________________________________________35

c. Freezing and thawing method _______________________________________36

2.1.2 Generation of OX-23 anti-factor H antibodies __________________________36

a. Hybridoma culture________________________________________________36

b. Production of conditioned media_____________________________________36

c. Purification _____________________________________________________37

d. Determination of protein concentration________________________________37

e. Titration________________________________________________________37

f. SDS-PAGE and Coomassie staining__________________________________38

2.1.3 Anti-A549 polyclonal antibody generation_____________________________39

a. Production of antibodies in NZW rabbits ______________________________39

b. Immunoreactivity assay____________________________________________40

2.1.4 Factor H purification______________________________________________40

a. Preparation of columns ____________________________________________40

b. Factor H purification ______________________________________________41

c. Factor H integrity ________________________________________________41

d. Factor H quantification by ELISA ___________________________________42

2.1.5 Alternative pathway hemolytic assay (APH50) _________________________43

2.1.6 Total RNA extraction and retrotranscription____________________________44

2.1.7 Protein extraction ________________________________________________46

2.1.8 Genotyping of C3 deficient mice ____________________________________46

2.2. Methods for the study of complement activity on cells ______________________47

2.2.1 Complement deposition on cells after activation of the alternative or classical

pathways _______________________________________________________47

2.2.2 Complement cytotoxicity after activation of the alternative or classical pathways __48

2.2.3 C5a quantification in plasma samples and bronchoalveolar lavages by ELISA _49

Page 26: Facultad de Ciencias - CORE

Index

2.2.4 Study of complement inhibitors in NSCLC and normal epithelial cells _______49

a. Determination of mRNA levels by quantitative PCR _____________________49

b. Factor H binding to the cell membrane ________________________________50

c. Detection of membrane-bound complement inhibitors ____________________50

d. Elimination of GPI-anchored proteins from the cell membrane by PI-PLC

treatment _______________________________________________________51

e. Effect of hypoxic conditions in the expression of complement inhibitors in

NSCLC cells ____________________________________________________51

2.3. Methods to study the in vitro effects of C5a ______________________________52

2.3.1 C5aR-1 expression _______________________________________________52

a. C5aR-1 mRNA expression _________________________________________52

b. C5aR-1 protein expression _________________________________________53

2.3.2 Treatment with C5a_______________________________________________53

2.3.3 C5a signaling activation ___________________________________________53

a. Calcium influx assay ______________________________________________53

b. ERK phosphorylation _____________________________________________54

c. NF-κB translocation to the nucleus___________________________________55

2.3.4 Effect of C5a on cell proliferation____________________________________55

2.3.5 Effect of C5a on cell cycle _________________________________________56

2.3.6 Effect of C5a on anchorage independent growth ________________________57

2.3.7 Effect of C5a on cell migration______________________________________57

2.3.8 Effect of C5a on HUVEC tube formation ______________________________58

2.3.9 Effect of C5a on cell adhesion to extracellular matrix proteins _____________58

2.3.10 Effect of C5a on integrin expression_________________________________59

2.3.11 Effect of C5a on MMP activity_____________________________________60

2.4. Mice models for the study of the influence of complement activation on lung tumor

growth in vivo______________________________________________________61

2.4.1 Syngeneic animal model of lung cancer _______________________________61

2.4.2 Tissue processing ________________________________________________63

a. Cell isolation from spleens _________________________________________63

b. Total RNA extraction from tumors and quantitative PCR _________________64

c. Fixation of tumors ________________________________________________64

d. Fixation of lungs _________________________________________________64

2.4.3 Study of angiogenesis by CD31 immunohistochemistry __________________64

2.5. Statistical analysis __________________________________________________65

RESULTS______________________________________________________ 67

1. COMPLEMENT ACTIVATION AND REGULATION ARE

INCREASED IN NON-SMALL CELL LUNG CANCER CELLS______ 69

1.1. Deposition of complement proteins after activation of the alternative pathway ___69

Page 27: Facultad de Ciencias - CORE

Index

1.2. Deposition of complement proteins after activation of the classical pathway _____70

1.3. Cell cytotoxicity due to complement activation by the alternative and classical

pathways _________________________________________________________73

1.4. Expression of complement inhibitors____________________________________74

1.5. Effect of hypoxia in the expression of complement inhibitors on NSCLC cells ___78

1.6. C5a levels in patients with NSCLC _____________________________________80

2. COMPLEMENT DEFICIENCY IMPAIRS TUMOR GROWTH ______ 81

2.1. Inhibition of tumor growth by C3 deficiency______________________________81

2.2. Inhibition of tumor growth by C5aR-1 blockade ___________________________83

3. COMPLEMENT ACTIVATION REGULATES ANTITUMOR IMMUNE

RESPONSE___________________________________________________ 85

3.1. Immune activation is induced in C3-deficient animals ______________________85

3.2. C5aR-1 blockade induces immune activation _____________________________87

3.3. C3 deficiency and C5aR-1 blockade reduces the expression of immunosuppressive

cytokines within the tumor____________________________________________89

4. EFFECT OF C5a IN THE PROMOTION OF ANGIOGENESIS ______ 91

4.1. Effect of C5a on HUVEC proliferation __________________________________91

4.2. Effect of C5a on HUVEC migration ____________________________________92

4.3. Effect of C5a on HUVEC tube formation ________________________________94

4.4. Decreased tumor angiogenesis in C3 deficient animals ______________________95

4.5. C5aR-1 blockade and C3 deficiency reduces bFGF expression in the tumor

microenvironment __________________________________________________96

5. EFFECT OF C5a ON NON-SMALL CELL LUNG CANCER CELLS__ 97

5.1. Expression of C5aR-1 in lung cancer cell lines ____________________________97

5.2. Activation of the C5a signaling pathway on NSCLC cells ___________________98

5.3. Effect of C5a on NSCLC cell proliferation ______________________________100

5.4. Effect of C5a in A549 migration ______________________________________101

5.5. Effect of C5a in NSCLC cell adhesion to extracellular matrix proteins ________102

5.5.1 Mechanisms involved in the regulation of cell adhesion to ECM proteins by C5a_103

5.5.2 Regulation of metalloprotease activity _______________________________104

DISCUSSION__________________________________________________ 107

1. THE DUAL ROLE OF COMPLEMENT ACTIVATION____________ 109

2. TUMOR CELLS ACTIVATE AND EFFICIENTLY REGULATE THE

COMPLEMENT CASCADE ___________________________________ 111

2.1. NSCLC cell lines spontaneously activate the complement system ____________111

2.2. NSCLC cells regulate complement dependent cytotoxicity__________________112

Page 28: Facultad de Ciencias - CORE

Index

3. EFFECT OF COMPLEMENT ACTIVATION IN THE TUMOR

MICROENVIRONMENT______________________________________ 116

3.1. Complement deficiency impairs tumor growth ___________________________116

3.2. Complement proteins and immunosuppression ___________________________120

3.3. Complement and angiogenesis________________________________________123

4. EFFECT OF C5a ON TUMOR CELLS __________________________ 125

5. COMPLEMENT IN HEALTH AND DISEASE____________________ 128

CONCLUSIONS _______________________________________________ 131

REFERENCES ________________________________________________ 135

Page 29: Facultad de Ciencias - CORE
Page 30: Facultad de Ciencias - CORE

INTRODUCTION

Page 31: Facultad de Ciencias - CORE
Page 32: Facultad de Ciencias - CORE

Introduction

3

1. LUNG CANCER

Cancer is a major public health problem in the western countries. For men, the

most common types of cancer are prostate, lung and colon; whereas, for woman, the

most common types are breast, lung and colon1. However, lung cancer is the leading

cause of cancer death for both men and women. More people die of lung cancer than of

colon, breast, and prostate cancers combined (Figure 1). This is a global fact, even

thought some progress has been made in the treatment of lung cancer in the last years,

using adjuvant, targeted and individualized chemotherapy. Only in the United States,

the American Cancer Society estimates for 2010 about 222,520 new cases of lung

cancer and about 157,300 deaths. In Europe, there were an estimated of 386,300 new

cases in 2006 (12.1% of overall new cancers diagnosed), ranking for the third in the list,

after breast and colorectal cancer; however, lung cancer, with an estimated 334,800

deaths (19.7% of total), was the most common cause of death from cancer2.

Figure 1. Estimated new cancer cases and deaths by sex in United States in 2009 for the ten leading cancer types. From Jemal et al.1

Page 33: Facultad de Ciencias - CORE

Introduction

4

1.1. Lung cancer classification

Lung cancer classification of the World Health Organization identifies two main

groups, with differences in their biological behavior and implications for treatment and

prognosis. These groups are: small cell lung carcinoma (SCLC), which accounts for

20% of lung cancers, and non-small cell lung carcinoma (NSCLC), which is the most

prevalent type and represents 80% of lung cancers.

NSCLC is further characterized by histology as adenocarcinoma, squamous cell

carcinoma, and large cell carcinoma. Adenocarcinomas represent about 40% of

NSCLC, are typically present in the lung periphery, may involve the pleura, and are

sometimes associated with scars. Bronchioalveolar carcinoma is a subtype of

adenocarcinoma which spreads along pre-exiting alveolar septae and does not invade

lung parenchyma. Squamous cell carcinomas, accounting for about 30% of NSCLC, are

typically more centrally located and posses features of squamous differentiation such as

keratin formation and intercellular bridges. Large cell carcinomas account for about

10% of all cases of NSCLC, may be central or peripheral, and exist in several subtypes.

NSCLC originates from epithelial cells and, even thought it is less aggressive than

SCLC, the detection usually takes place once the tumor has spread and metastases at

different points already exist3.

SCLC tends to arise in the larger airways and grows rapidly, becoming quite

large. The small cells contain dense neurosecretory granules, so are known to have an

endocrine origin. While initially this type of lung cancer is more sensitive to

chemotherapy, it ultimately carries a worse prognosis and is often metastatic at

presentation4.

1.2. Lung cancer staging

NSCLC is currently staged according to the T (extent of the primary tumor), N

(extent of involvement of regional lymph nodal disease), and M (extent of spread to

distant sites) scheme. The purpose of a staging system for any malignant disease is

threefold. First, it provides an estimate of tumor burden and extent of spread; second, it

provides prognostic information that can be applied to individual patients; and third, an

effective staging system is able to direct therapy. A recent universally stage

classification for NSCLC has been accepted by the Union Internationale Contre le

Cancer (UICC) and the American Joint Committee on Cancer (AJCC)5.

Page 34: Facultad de Ciencias - CORE

Introduction

5

More than 85 percent of people who are diagnosed with lung cancer today are

diagnosed in a late stage, when the disease is more difficult, and sometimes impossible,

to cure (Table 1). Therefore, an obvious strategy to reduce mortality would be the early

detection of the disease. The International Early Lung Cancer Action Program (I-

ELCAP) has shown that 85% of lung cancers can be found in the earliest, most curable

stage when an annual CT screening program is followed. If treated promptly with

surgery, the cure rate of these early stage patients is 92%6. Moreover, any patient

diagnosed with lung cancer as a result of I-ELCAP annual CT screening has an

estimated cure rate of 80%, regardless of stage and type of treatment (www.ielcap.org).

Recently, the National Lung Screening Trial (NLST), a randomized trial, found 20%

fewer lung cancer deaths among participants screened with low-dose helical CT

(www.cancer.gov). In any case, primary prevention services are still the more crucial

strategy to reduce lung cancer mortality. Besides, more research on the biological

mechanisms involved in the disease would allow the identification of high risk

individuals, the characterization of biomarkers that may signal the presence of lung

cancer, and the development of more efficient therapies.

Table 1. Lung cancer staging, and the related prognosis, according to the TNM system.*

*Modified from Detterbeck et al.5 **Median survival time (MST) in months, and 5-year overall survival.

Stage T N M PROGNOSIS** MST (5 years)

Ia T1a,b N0 M0 60-119 50-73%

Ib T2a N0 M0 43-81 43-56%

IIa T1a,b

T2a

T2b

N1

N1

N0

M0

M0

M0

34-49 36-46%

IIb T2b

T3

N1

N0

M0

M0 18-31 25-36%

IIIa T1-3

T3

T4

N2

N1

N0,1

M0

M0

M0

14-22 19-24%

IIIb T4

T1-4

N2

N3

M0

M0 10-13 7-9%

IV Tany Nany M1a,b 6-17 2-13%

Page 35: Facultad de Ciencias - CORE

Introduction

6

1.3. Lung cancer etiology

It is estimated that about 80% of lung cancer deaths are related to tobacco

smoking7. Apart from smoking, other associations have been seem with lung cancer.

For example, individuals with a history of chronic obstructive pulmonary disease8, or

even individuals with emphysema but without airway obstruction, are associated with

an increased risk for lung cancer9. Other contributing factors may include exposure to

radon, asbestos, arsenic, chromium, nickel and air pollution10. There is also an

increasing interest in the identification of genetic variations associated with increased

risk of lung cancer. In 2008, three independent groups simultaneously identified

common inherited genetic variations in chromosome 15, containing nicotinic

acetylcholine receptors, associated with increased susceptibility to lung cancer11-13.

2. THE COMPLEMENT SYSTEM

The complement system has been recognized classically as a central part of the

innate immune response, which develops a first defense against microbes and unwanted

host molecules. Its physiological functions are: defending against pyogenic bacterial

infection, bridging innate and adaptative immunity, and disposing of immune

complexes and the products of inflammatory injury14. All these activities are mediated

by more than 30 circulating or cell-surface-bound proteins. These proteins can function

as enzymes, substrates or regulators, keeping a well-balanced activation and inhibition

of the system. A very comprehensive review has recently been published that

summarizes the function and structure of complement proteins and describes in detail

the multiple roles of complement in homeostasis15.

Classically there are three established mechanisms of complement activation,

known as classical, lectin and alternative pathways (Figure 2). Each one differs from

the others in the initial steps, the proteolytic phase, and they converge in the cleavage of

the central molecule of the pathway, C3, which generates its active fragments C3a and

C3b. Complement is also activated by additional bypass pathways that act

independently of the classical and the alternative convertases (see section 2.1.4.). The

non-proteolytic phase starts after the cleavages of C3 and C5, when the following

components ensemble on the membrane and form a multiprotein pore complex (the

membrane attack complex, MAC) which eventually leads to cell lysis. A series of

Page 36: Facultad de Ciencias - CORE

Introduction

7

complement receptors on leukocytes mediate the recognition of opsonized cells, which

results in phagocytosis and the stimulation of the adaptive immune system. Finally, the

anaphylatoxins C3a and C5a are released during complement activation and trigger a

range of chemotactic and pro-inflammatory responses, such as the recruitment of

inflammatory cells and the increase in microvasculature permeability.

MBL

C1 complex

Ab

C4

C4b C2

C4b2b

C3

C3b

C4b2b3b C3bBb3b

C3C3H2O

C3H2O& fB

fD

C3H2OfBC3

C3b & fB

C3C3bBb &

properdin

C5C5

C5b

C6C7 C8

nC9MAC

C3a

C3a

C5a

fD

LECTIN PATHWAY

CLASSICAL PATHWAY

ALTERNATIVE PATHWAY

MBL

C1 complex

Ab

C4

C4b C2

C4b2b

C3

C3b

C4b2b3b C3bBb3b

C3C3H2O

C3H2O& fB

fD

C3H2OfBC3

C3b & fB

C3C3bBb &

properdin

C5C5

C5b

C6C7 C8

nC9MAC

C3a

C3a

C5a

fD

LECTIN PATHWAY

CLASSICAL PATHWAY

ALTERNATIVE PATHWAY

Figure 2. Drawing showing the cascade of events during the activation of the complement system.

Excessive complement activation on self tissue has severe effects and can

promote the development of various diseases. For this reason, a range of soluble and

cell-surface-bound inhibitors ensures that any action of complement on host cells is

either prevented or actively inhibited.

Page 37: Facultad de Ciencias - CORE

Introduction

8

2.1. Complement activation

2.1.1 The classical pathway

The first component of the classical pathway is a complex formed by three

proteins: C1q, C1s and C1r. The activation takes place after the hexameric C1q

recognizes the antibody constant regions of µ chains (IgM) and some γ chains (IgG)

bound to the target antigen16. In addition, it has been shown more recently that the

classical pathway can also be activated in the absence of antibodies, as C1q recognizes

endogenous ligands such as dying cells17, extracellular matrix proteins18, pentraxins19,

amyloid deposits20, prions21 and DNA22. Binding of C1q activates C1s and C1r. C1s is a

serine protease which cleaves two serum proteins, C4 and C2. The cleavage of C4

generates two fragments: C4b, which keeps bound covalently to the cell surface through

a thioester bond, and C4a, a soluble anaphylatoxin that diffuses away. The third

component, complement C2, binds to C4b and becomes a target for C1s as well. The

cleavage of C2 generates also two fragments, C2b which keeps bound to C4b, and C2a.

C4bC2b forms the classical C3 convertase on the cell surface; C2b in the convertase

complex acts as a serine protease that cleaves C3 in C3b and C3a. C3b binds covalently

to the cell membrane due to its thioester bond, and joins to the C3 convertase, forming

the classical C5 convertase, C4bC2bC3b. In addition, C3b acts as an opsonin by

covering the cell surface. The soluble C3a is the second anaphylatoxin generated in the

classical pathway. The activation of C5 occurs in the same manner, after C5 binding to

its convertase, it is cleaved in C5a, the most potent anaphylatoxin, and C5b, which starts

the second phase of complement activation. This stage results in directed, non-

enzymatic assembly of the terminal pathway components. First, C6 binds to C5b, and

then C7 binds to both and inserts into the cell membrane. The subsequent binding of C8

causes a deeper penetration of the complex into the membrane. All the conformational

changes that occur during the assembly induce the polymerization of 15-19 molecules

of C9 into the cell membrane. This action generates a 100 Å pore on the membrane, as

seen by electron microscopy23. The structure formed is known as the terminal

complement complex (TCC) or the membrane attack complex (MAC), and is

responsible for the final consequence of complement activation, the osmotic lysis of the

cell24.

Page 38: Facultad de Ciencias - CORE

Introduction

9

2.1.2 The lectin pathway

The lectin pathway is analogous to the classical pathway. This pathway is

activated by homologous proteins to C1q: mannose-binding lectin (MBL) and ficolins.

These proteins recognize specific carbohydrates on pathogens’ surface, such as

mannose and N-acetyl-glucosamine (GlcNAc). After their binding to the carbohydrates,

MBL-associated serine proteases (MASP-1 and MASP-2) function in an analogous

manner to C1r and C1s25, cleaving the complement components C4 and C2 to form the

C3 convertase C4bC2b, which is common to both the lectin and classical pathway

activation routes26.

2.1.3 The alternative pathway

The alternative pathway is mechanistically distinct from the classical and lectin

pathways. It is antibody-independent and provides an initial line of innate immune

defense, as it is initiated by the low-level and spontaneous hydrolysis of C3 on

activating surfaces. The spontaneous cleavage of C3, also known as “the tickover of

C3”, forms C3(H2O), which contains an activated thioester bond. C3(H2O) can bind to

factor B, in turn allowing its cleavage by factor D into Bb and Ba, forming the initial

alternative C3 convertase, C3(H2O)Bb27. This complex starts to convert C3 into C3b

and C3a. In most cases this C3b is rapidly inactivated. However, in complement

activating surfaces, the C3b generated can bind to them and associate with factor B. In

the last complex, C3bB, factor B is again a substrate for the factor D cleavage, forming

the predominant alternative C3 convertase, C3bBb. The stability of this convertase is

enhanced by the binding of properdin, helping to amplify the alternative pathway

activation28. The fragment Bb on the C3 convertase plays an analogous role to its

homologous C2b in the classical convertase. Bb cleaves C3 and forms an amplification

loop, generating more C3b that, on one hand, can create new alternative C3 convertases,

and on the other hand, is necessary to form the C5 convertase (C3bBbC3b). After C5

cleavage into C5a and C5b, the pathway continues with the non-enzymatic assembly of

the terminal pathway components.

2.1.4 Alternative routes of complement activation

Apart from the three major activation mechanisms, there are several bypass routes

that have been shown to directly trigger complement response at various stages. The

lectin pathway can be activated directly by the binding of MBL to natural IgM bound to

Page 39: Facultad de Ciencias - CORE

Introduction

10

ischemic antigens in endothelial cells after ischemia/reperfusion injury29-30. In the

absent of C2/C4, but in the present of alternative pathway components, it is possible

that antigen-antibody complexes or oligosaccharides activate C1 or MBL,

respectively31-32. It has been also shown that anaphylatoxins can be generated due to the

action of certain proteases. C3 can be cleaved and activated by extrinsic proteases, such

as thrombin or kallikrein, pointing to a cross-talk between the complement system and

the coagulation cascade33. C5 can be cleaved by thrombin, bypassing C334. C3a and C5a

can be generated directly from C3 and C5 by proteases found in the allergenic feces

produced by dust mites35-36. And finally, C5 can be cleaved by silica37 and asbestos

fibers38, by mechanisms involving the generation of free radicals and kallikrein

activation39.

2.2. Complement regulation

Regulation of complement is of critical importance for the homeostasis of the

organism. In order to avoid tissue damage, control proteins regulate complement at

three main levels: they can inhibit the protease activity of the proteins involved in the

activation cascade, facilitate the decay and destruction of convertases, and control the

formation of MAC (Figure 3). Usually complement regulators are grouped into two

groups, soluble proteins and membrane-bound proteins (known as membrane

complement regulatory proteins or mCRP).

Regulators share a varying number of a repeating motif termed complement control

protein (CCP) repeat, also known as short consensus repeat (SCR) or sushi domain.

CCP domain is characterized by its length (each unit contains approximately 60 amino

acids), its globular structure, and a frame of conserved residues that includes four

invariant cysteines, an almost invariant tryptophan and highly conserved prolines,

glycines and hydrophobic residues40. They are thought to play a role in binding to C3,

C4 and their breakdown products.

Page 40: Facultad de Ciencias - CORE

Introduction

11

C1 INH

Factor H

FHL-1

C4BP

CD31

CD55

CD46

ClusterinVitronectin

CD59

Factor I

MBL

C1

C4b

C3bBbC3b

iC4b

iC3b

MAC

C4b2b

C1 INH

Factor H

FHL-1

C4BP

CD31

CD55

CD46

ClusterinVitronectin

CD59

Factor I

MBL

C1

C4b

C3bBbC3b

iC4b

iC3b

MAC

C4b2b

Figure 3. Drawing of the main complement inhibitors, soluble proteins and mCRPs. Red lines represent inhibitory activity (when ending in a bar) or accelerated decay activity (when ending in a square); green lines represent cofactor activity (when ending in a square) or protease activity (when ending in an arrow head).

2.2.1 Soluble regulators of the complement system

a. C1 inhibitor

C1 inhibitor (C1 INH) is a member of the serpin family of protease inhibitors,

although it presents unique features that differentiate C1 inhibitor from other serpins41.

The protease inhibition carried out by C1 inhibitor is the most important activity

of this protein. This protease inactivates C1r, C1s and MASP2, resulting in the

inhibition of classical and lectin complement pathways. In addition, C1 INH inactivates

a variety of proteases outside the complement system: intrinsic pathway proteases

(factor XII, plasma kallikrein, factor XI), thrombin and fibrinolytic proteases (plasmin,

tissue plasminogen activator)42.

Page 41: Facultad de Ciencias - CORE

Introduction

12

b. C4-binding protein

Human C4-binding protein (C4BP) is a heterogeneous oligomeric protein present

in plasma in three isoforms, with different subunit composition. The major isoform is

composed of seven identical 75 kDa alpha-chains and a unique 40 kDa beta-chain, that

are held together by disulfide bridges. Alpha and beta-chains are formed by eight and

three SCR modules, respectively.

C4BP mostly inhibits the classical complement pathway. C4BP, after binding to

C4b, inhibits complement by three different ways: prevents the assembly of the C3

convertase, accelerates the decay of the classical C3 and C5 convertases, and functions

as a cofactor in the factor I-mediated inactivation of C4b. C4BP also plays a role as a

regulator of the alternative pathway by acting as a cofactor of factor I in the cleavage of

C3b, but only in the fluid phase43.

Outside the complement system, C4BP binds to the serum amyloid P component

(SAP) and to the anticoagulant protein S, again pointing to a cross-link between

complement and coagulation systems44.

c. Factor H and factor H-like protein 1

Factor H is a 150 kDa glycoprotein composed of 20 SCRs. In addition, alternative

splicing of factor H mRNA yields a 42 kDa protein, named factor H-like protein 1

(FHL-1) or reconectin. FHL-1 is identical in sequence to the seven N-terminal SCRs of

factor H, with four additional amino acids added at the C-terminal end. FHL-1 shares

the complement inhibitory activities of factor H.

Factor H is mostly known as an inhibitor of the alternative pathway. Through its

binding to C3b, factor H competes with factor B in the formation of the C3 and C5

convertases, displaces the Bb subunit from the convertases, and acts as a cofactor for

factor I in the cleavage of C3b45.

Several recent studies have described the association of genetic variations in

complement factor H with different diseases. Mutations or polymorphisms that alter the

binding of factor H to C3b and polyanions are associated with atypical hemolytic

uremic syndrome (aHUS), whereas mutations that disrupt the plasma activities of factor

H, leading to unrestricted activation of the alternative pathway, are associated with

membranoproliferative glomerulonephritis type II (MPGN2)46. The most studied

polymorphism at the factor H locus is rs1061170, which causes a Tyr402His amino acid

substitution in SCR7. Independent studies have shown that the allele 402His confers a

Page 42: Facultad de Ciencias - CORE

Introduction

13

significantly increased risk to age-related macular degeneration (AMD) in many

different populations47-50 possibly by altering the binding of SCR to different

glycosaminoglycans and retinal pigment epithelial cells46.

In addition, factor H displays functions outside the complement system. It serves

as a chemotactic factor and a secretagogue of IL-1β for monocytes, and as an adhesion

protein for neutrophils51-52.

Five complement factor H-related proteins (CFHR) have been identified: CFHR-1

to -5. They are coded by five genes closely linked to the factor H gene, and are

composed of SCRs with different degrees of identity with SCRs in factor H53. Like

factor H, CFHRs are mostly synthesized in the liver, but their concentrations are much

lower than that of factor H, and their functional properties are not well defined. Only

FHR-5 displays a weak cofactor activity in cleavage of C3b by factor I. FHR-1 binds to

C3b and may have a possible role in complement regulation. FHR-3 binds to C3b, C3d

and heparin, and FHR-4 to C3b, which also supports the role of these proteins in the

complement pathway (reviewed by Serka and Zipfel54).

d. Vitronectin

Vitronectin (S protein, S40) exists as a 75-kDa protein in the extracellular matrix

(ECM), and as two truncated forms in plasma, which are held together by a disulfide

bond. Vitronectin inhibits the insertion of C5-7 at its membrane-binding site. The fluid

phase sC5b-7 is still able to bind C8 and C9 to form soluble non-lytic C5b-8 and C5b-9

complexes. It has also been shown that vitronectin in solution directly inhibits C9

polymerization, and hence indirectly can block pore formation55.

In addition, vitronectin is attached to the extracellular matrix via its collagen

binding- or heparin binding-domains where provides a regulatory link between cell

adhesion and physiological proteolysis (reviewed by Schavartz et al.56).

e. Clusterin

Clusterin (apolipoprotein J) is a ubiquitously expressed protein. Structurally is a

heterodimer linked by a unique five disulfide bond motif. It functions similar to

vitronectin in complement inhibition by binding to the C5b-7 complexes and preventing

C9 polymerization57. Appart from the complement inhibitory function, clusterin plays

other physiologic functions that depend on its final maturation and localization. It has

been shown that intracellular forms of the protein display direct or indirectly

Page 43: Facultad de Ciencias - CORE

Introduction

14

pro/antiapoptotic activity and act as a negative regulator of NF-κB. Clusterin can also

modulate differentiation and regulate the production of major proinflammatory

cytokines such as TNF-α and IL-6. For all these functions, it has recently been related to

inflammation and autoimmunity58.

2.2.2 Membrane complement regulatory proteins

a. Complement receptor type 1

Complement receptor type 1 (CR1, CD35) is comprised of a linear array of 30

SCR units in its extracellular region, and is found in erythrocytes, neutrophils,

eosinophils, monocytes, follicular dendritic cells, glomerular podocytes, B lymphocytes

and some T lymphocytes59. CR1 regulates complement activation acting as a cofactor

for factor I-mediated cleavage of C3b and C4b, and accelerating the decay of the

classical and alternative convertases60.

b. Membrane cofactor protein

Membrane cofactor protein (MCP, CD46) is expressed as four common isoforms,

all of them composed of four SCR units. It is expressed in most cells but erythrocytes. It

acts as a cofactor of factor I for the C3b and C4b cleavage61.

In addition to its function as complement inhibitor, CD46 has been implicated in

the regulation of T cell-induced inflammation62 and the generation of T regulatory

cells63. CD46 also serves as receptor for several human viruses and bacteria: measles

virus human herpesvirus 6, adenovirus of different serotypes, Streptococcus pyogenes

and pathogenic Neisseria (reviewed by Cattaneo64).

c. Decay accelerating factor

Decay accelerating factor (DAF, CD55) is a glycoprotein composed of four SCR

domains which is anchored to the membrane by a glycosylphosphatidylinositol (GPI)

anchor. It is present in all blood elements and most other cell types65. DAF accelerates

the decay of the classical and alternative C3 and C5 convertases66.

CD55 binds to its ligand CD97, expressed on macrophages, granulocytes, and

dendritic cells, and is rapidly up-regulated in activated T and B cells. Through this

binding, CD55 might simultaneously regulate both the innate and adaptive immune

responses67.

Page 44: Facultad de Ciencias - CORE

Introduction

15

d. CD59

CD59 (protectin) is a compact single chain of 77 amino acids with a disc-shaped

structure, and, like CD55, is attached to the cell membrane by a GPI anchor. It is widely

expressed, present on all circulating cells, vascular endothelium, epithelium and most

other cell types. CD59 binds C8 in the forming MAC and inhibits the insertion of C9

into the lipid bilayer68. Alternative roles of CD59, related to its GPI anchor signaling

properties, have also been demonstrated in cell types such as T cells, natural killer cells

and B cells69.

2.3. Biological effects of complement activation

2.3.1 Cell lysis by the membrane attack complex

Complement was first recognized because of its capacity to lyse cells, in

particular bacteria. It has been shown that a single functional MAC in the membrane of

an erythrocyte is sufficient to lyse it70. The formed pore permits entry of water into the

cell, driven by the osmotic gradient, causing the cell to swell and burst. However,

nucleated cells are more resistant to this cytotoxic effect. Not only do they need a higher

number of pores on the cell surface, but also other factors are important. One essential

factor is the calcium influx into the cell once a number of MAC is formed. An increase

of intracellular calcium concentration causes a loss of mitochondrial transmembrane

potential resulting in an energy crisis71. And then, the loss of ATP and its precursors

through the MAC leads to cell necrosis72.

Apart from causing cell necrosis, MAC can induce other responses in the cell.

MAC can activate immune cells to release inflammatory molecules, increase the

resistance of cells to further lytic attack, drive cells to proliferate, and make cells more

resistant to apoptosis73.

2.3.2 Opsonization of the cell by C3b and its related fragments

All complement pathways lead to the central and initial step in complement

opsonization, the conversion of the component C3 to C3b. The nascent C3b molecule

can lead to complement amplification through the actions of factors B and D, but can be

also inactivated by factor I and its cofactors (MCP, CR1, CR2 and factor H). Initial

inactivation of C3b is mediated by factor I, which catalyzes the proteolysis of two

peptide bonds in the α' polypeptide chain of C3b. The resulting products are the

Page 45: Facultad de Ciencias - CORE

Introduction

16

membrane bound iC3b and a small peptide, C3f, which is released from the molecule.

The third cleavage catalyzed by factor I generates the inert C3c and C3d-g, the former is

released and the later is kept on the cell membrane. Although further complement

amplification is abolished, recognition of C3b and its fragments by complement

receptors on cells promotes phagocytosis74.

Complement receptors are divided in three recognized gene families: the SCR

family members CR1 and CR2, the β2 integrin family members CR3 and CR4, and the

immunoglobulin Ig-superfamily member CRIg.

CR1 (CD35) is expressed in the majority of peripheral blood cells and recognizes

C3b, C4b, iC3b, C3dg, C1q and mannose-binding protein75-76. The main function of

CR1 is to capture immune complexes (IC) on erythrocytes for transport and clearance

by the liver77. Apart from this role in IC elimination, CR1 promotes the secretion of

proinflammatory molecules, such as interleukin (IL)-1α, IL-1β, and prostaglandins; has

a role in antigen presentation to B cells; and is an inhibitor of both classical and

alternative pathways, due to its decay-accelerating activity and cofactor activity for

C3b/C4b cleavage78-79.

CR2 (CD21) is an evolutionary homologue of CR1, but only binds to iC3b, C3d

and C3dg. On B cells, CD21 forms a co-receptor with CD19 and CD81. Binding of the

co-receptor to the B cell antigen receptor (BCR) lowers the threshold for B cell

activation80. This fact is due to the uptake of C3d-coated antigens by B cells81.

CR3 (CD11b/CD18) and CR4 (CD11c/CD18) are expressed by leukocytes and

stimulate phagocytosis when bound to iC3b. In addition, they perform functions in

leukocyte trafficking, adhesion, migration, and costimulation, which have important

consequences in the defense of the host against pathogenic invasion82.

CRIg has been identified recently as a complement receptor. It is expressed on a

restricted subset of tissue-resident macrophages. CRIg may play an important role in

phagocytosis83.

2.3.3 Biological effects mediated by anaphylatoxins

During complement activation, soluble active fragments of approximately 9 kDa

(74-77 residues) are released from C4, C3 and C5. These molecules, named C4a, C3a,

and C5a, were called anaphylatoxins as they are potent multifunctional pro-

inflammatory molecules. In addition to complement activation, C3a and C5a can be

directly generated by proteases unrelated to the complement cascade (see section

Page 46: Facultad de Ciencias - CORE

Introduction

17

2.1.4.). Anaphylatoxins are evolutionarily related to one another, sharing a relatively

high degree of homology: C5a shares with C3a 36% amino acid identity, and C4a

shares 33% and 25% homology with C5a and C3a, respectively84. They also have some

overlapping functions in the generation of the immune response.

Anaphylatoxins play their functions after the binding to their receptors, which

belong to the superfamily of G-protein-coupled receptors (GPCR). Anaphylatoxin

receptors share high sequence homology85 and are closely related to other chemotactic

receptors such as the N-formyl-methionine-leucine-phenylalanine (fMLP) receptor,

ChemR23 and the chemokine receptors CXCR1 and CXCR286. Even though their

similarity, anaphylatoxin receptors differ in ligand specificity, signal transduction

capacity and function. The anaphylatoxin receptors comprise C3aR for C3a, and C5aR

and C5L2 for C5a. To date, no specific receptor has been found for C4a, which was

originally reported to bind to the same receptor as C3a87. For C5a binding, the first

recognized receptor was C5aR-188, also known as CD88; but more recently the orphan

receptor GPR77 was identified as a second C5a receptor and was called C5a-like

receptor 2 (C5L2)89. Both are seven-transmembrane receptors that share a 35%

homology. However, C5aR is a classical G protein-coupled receptor, while C5L2 is an

enigmatic receptor deficient in G protein coupling. This fact, together with an unknown

pathway for C5L2 after C5a binding, led to suggest that C5L2 is a default receptor that

attenuates C5a biological responses by competing with C5aR-1. Nevertheless, this

definition for C5L2 seems to be incorrect, at least in part, because current contrasting

results point to anti-inflammatory functions for C5L290 or a positive regulatory activity

of C5aR91.

Initially, C3aR-1 and C5aR-1 expression was reported only on myeloid cells, but,

in the recent years, there have been numerous reports demonstrating their expression in

a wide variety of non-myeloid cell types92. C3aR is expressed in inflamed human

central nervous system93, endothelial cells94, epithelial cells, smooth muscle cells,

submucosal and parenchymal vessels of the lung from asthma patients95, and activated

human T cells96. C5aR is expressed in a broad range of non-immune cells: endothelial

cells, neurons, astrocytes and microglia, cells from kidney, lung, liver, spleen, intestine,

skin and heart97. C5L2 seems to be frequently co-expressed with C5aR in most cells or

tissues98-99. Activities of anaphylatoxins are related to the cell types which express their

receptors. Classically, they have been known to increase vascular permeability, promote

smooth muscle contraction, induce leukocyte recruitment, increase chemotaxis,

Page 47: Facultad de Ciencias - CORE

Introduction

18

migration and phagocytosis in white blood-cell, and promote the production and release

of other inflammatory mediators (e.g., histamine)100. Apart from their pro-inflammatory

properties, anaphylatoxins have also been implicated in tissue regeneration and tissue

fibrosis101, and in brain development102. In summary, they are recognized as pleiotropic

molecules that can modulate the activity of many cell types, with a broad range of

biological functions, both inside and outside of the immune system.

The potency and duration of these activities differ among the three

anaphylatoxins. C5a has been shown to be considerably the most potent, whereas C4a is

the weakest in inducing biological responses. In addition, anaphylatoxin activity, in the

circulation and in tissues, is tightly controlled by carboxypeptidases, which rapidly

cleave off a C-terminal arginine residue, generating the desArg forms103. C5a-desArg

binds with 100-fold less affinity to C5aR, but with the same affinity to C5L2. C3a-

desArg is devoid of any pro-inflammatory activity104. The clearance of C5a-desArg

from the body is achieved after its binding to C5aR on leukocytes and other cells105, or

even more effectively after binding to C5L2, as it is internalized, retained and degraded.

This function of C5L2 suggests that a major function of this receptor is to remove active

complement fragments from the extracellular environment106.

In the recent years the interest of C5a in cancer progression has increased due to

the connection between this anaphylatoxin and the immunosuppression of tumors107.

3. COMPLEMENT AND CANCER

3.1. Complement in the immune surveillance against tumors

Once a threatening body is recognized by the complement system, the activating

steps are followed, generating an inflammatory reaction, the opsonization of pathogens,

and eventually the killing of some organisms. Considering this classical role of

complement, a similar scenario could be considered for the control of tumor growth.

During carcinogenesis, cancer cells acquire several sequential genetic and epigenetic

abnormalities that result in essential alterations in the cell physiology. These alterations

dictate malignant growth and produce changes in cell morphology, generating many

tumor-associated antigens that distinguish malignant cells from their normal

counterparts; for example, a change in glycosylation phenotype is considered a hallmark

of cancer cells108. Those changes can induce the recognition of malignant cells by the

Page 48: Facultad de Ciencias - CORE

Introduction

19

immune system109. In fact, in cancer patients, complement activation, with subsequent

deposition of complement components on tumor tissue, has been demonstrated. Thus,

deposits of C3, C4 and C5b-9 were found in breast110, and papillary thyroid

carcinoma111. In lung tumors, only C3b, but not MAC deposition, was detected by

immunohistochemistry112. Elevated levels of C3a and soluble C5b-9 were present in the

intraperitoneal ascitic fluid of ovarian cancer patients113. The MBL complement

activation pathway was significantly increased in patients with colorectal cancer

compared with healthy persons114. Higher complement hemolytic activity and C3 levels

were observed in serum samples from children with neuroblastoma115 and, similarly,

elevated complement levels were reported in patients with carcinomas of the digestive

tract116 and brain tumors117. In summary, although there are studies reporting normal

total complement and C3 levels in patients with breast, gastric and colorectal

carcinomas118, most in vivo observations suggest that cancer cells activate the

autologous complement system.

Therefore, complement recognition of cancer cells may be an element of the

immune surveillance theory, which supports that the immune system surveys the body

for tumor-associated antigens, taking part of the elimination of many or most tumors119.

However, it is clinically evident that this system sometimes fails to protect the organism

from growing tumors. The principal reason is the development of active mechanisms to

escape immune recognition or resist immune attack.

3.2. Complement regulation on cancer cells

There is sufficient basis to propose that certain cancers activate, directly or

indirectly, the complement system. However, although neoplastic transformation may

be accompanied by an increased capacity to activate complement, cancer cells seem to

develop a collection of mechanisms to resist complement attack. Those mechanisms can

be divided into two categories, extracellular and intracellular mechanisms (reviewed by

Jurianz et al.120). Extracellular protection is provided by:

(1) Expression of mCRPs on the cell surface. Several studies have demonstrated

the expression of mCRPs in primary tumors and cell lines. With the exception of CR1,

most cancers, independent of their tissue origin, express at least two, if not three,

mCRPs. In addition, it has been shown that mCRPs are overexpressed on tumor cells

(reviewed by Fishelson et al.121).

Page 49: Facultad de Ciencias - CORE

Introduction

20

(2) Release of soluble complement inhibitors, C1 inhibitor, factor H, factor I, or

clusterin, and binding of such inhibitors to the cell membrane; where they regulate

complement activation and subsequent complement dependent cytotoxicity (CDC). In

this sense, our group demonstrated that NSCLC cell lines express and release to the

extracellular milieu factor H and FHL-1122; in addition, downregulation of factor H in

one of those cell lines reduces its growth in vivo123. In a different study, we

demonstrated that NSCLC cell lines express functional factor I and C4BP, which

support efficiently the cleavage of C3b and C4b in vitro124. Another similar mechanism

is the production and release of soluble forms of mCRPs, such as sCD46, sCD55 or

sCD59, by alternative splicing or enzymatic cleavage of the membrane anchoraged

form.

(3) Secretion of chondroitin sulfate proteoglycans by malignant B lymphocytes

that resemble, in structure and function, the C1q inhibitor 125.

(4) Release of ecto-proteases by tumor cells that cleave complement components,

mainly C3126.

(5) Expression of ecto-protein kinases on the cell membrane. One example is the

casein kinase 2 (CK2), an ubiquitous and constitutively expressed protein kinase. It has

been demonstrated that CK2 expressed on the surface of human leukemia cell lines

phosphorylates several exogenous substrates, including the complement C9 protein127.

Phosphorylation of C9 may protect tumor cells from complement, either by inhibiting

MAC formation or by leading to the production of an inactive or unstable MAC. It has

been shown that blocking CK2 with selective inhibitors enhances killing of

lymphoblastoid cells due to the classical pathway activation by Rituximab128, an

antibody against the protein CD20 used in the treatment of lymphomas and leukemias.

Intracellular protection is provided by sublytic doses of MAC. Surprisingly, the

MAC itself is one of the most potent mechanisms to increase resistance to complement

attack. Its insertion on the cell membrane causes a variety of biological effects, such as

entrance into cell cycle, resistance to apoptosis, expression of adhesion molecules or

augmentation of complement resistance (reviewed by Morgan129). The mechanisms

responsible for this protection are poorly understood, but may involve increase in

calcium intracellular concentrations, PKC and MAPK activation, and protein and RNA

synthesis130. It is possible that those mechanisms are orchestrated to remove MAC from

the cell surface by external vesiculation or internalization.

Page 50: Facultad de Ciencias - CORE

Introduction

21

3.3. Complement in the immunotherapy of tumors

Tumor specific and tumor associated-antigens can be used as targets for

monoclonal antibody (mAb) immunotherapy. The number of mAbs that have been used

in the clinic as therapeutic drugs has been increasing since the first mAb, Rituximab,

was used for the treatment of lymphomas131. Those mAbs target tumor-specific and

tumor-associated antigens and block important cancer activities. In addition, most of

them are of the IgG1 isotype, which effectively mediates Fc domain–based functions,

such as antibody-dependent cellular cytotoxicity (ADCC) and complement fixation. It is

more successful to use a mAb that combines different mechanisms to direct a harmful

effect on tumor cells. For that reason, researchers are motivated to try to increase the

activation of complement on tumors using mAbs that already block another function on

the cell. However, the resistance of tumor cells to complement activation has to be

overcome.

Different strategies have been developed to overcome protection by complement

regulators: use of mCRP blocking antibodies in combination with anticancer

complement-fixing antibodies121; down-regulation of mCRP expression using

cytokines132; and removal of CD55 or CD59 from the cell surface using GPI-

phospholipase C133-134. However, the mentioned mechanisms are not specific for tumor

cells, and the aim now is to achieve an inhibitory effect of complement regulators

limited to the tumor. It seems that blockade of mCRPs is the most promising way to

achieve it. Some strategies include the use of a biotin-avidin system135, to neutralize

selectively complement regulators using antibodies136, and to use therapeutic bispecific

mAbs that target a tumor antigen and simultaneously block a major complement

regulatory protein137.

3.4. Complement, inflammation and cancer

Since the beginning of the 19th century, with the Virchow’s theory, the link

between cancer and inflammation was perceived. Virchow observed that chronic

inflammation establishes an environment that promotes the initiation and growth of

malignancy138. Since then, epidemiological studies have shown the growing evidence

that chronic inflammation predisposes individuals to various types of cancer. It is

estimated that 15–20% of all deaths due to cancer worldwide are attributable to

inflammatory processes139. Different triggers of chronic inflammation are related to the

Page 51: Facultad de Ciencias - CORE

Introduction

22

increased risk of malignancy, such as microbial infections, autoimmune diseases,

chemical and physical agents, and inflammatory reactions of uncertain etiology

(summarized in Table 2). In addition, the usage of non-steroidal anti-inflammatory

drugs may protect against various tumors140.

Table 2. Some associations between inflammation and cancer risk.

*Adapted from Balkwill and Mantovani138.

An inflammatory component is also present in the microenvironment of tumors

that are not epidemiologically related to inflammation. Within the microenvironment of

tumors, there are several inflammatory elements that feed tumor cells in terms of

inducing their proliferation and survival, promoting angiogenesis and metastasis,

evading adaptive immunity, and reducing response to hormones and chemotherapeutic

agents. Recently, it has been proposed that an inflammatory microenvironment might be

summed up to the six hallmarks of cancer proposed by Hanahan and Weinberg in

2000141 (Figure 4).

Malignancy Inflammatory stimulus/condition

Bladder Schistosomiasis

Cervical Papillomavirus

Ovarian Pelvic inflammatory disease/talc/tissue remodeling

Gastric H pylori induced gastritis

MALT lymphoma H pylori

Esophageal Barrett’s metaplasia

Colorectal Inflammatory bowel disease

Hepatocellular Hepatitis virus (B and C)

Bronchial Silica, asbestos, cigarette smoke

Mesothelioma Asbestos

Kaposi’s Sarcoma human herpesvirus type 8

Page 52: Facultad de Ciencias - CORE

Introduction

23

Figure 4. Inflammation as the seventh hallmark of cancer from Colotta et al.141

The main features of cancer-related inflammation include the infiltration of white

blood cells, prominently tumor-associated macrophages (TAMs) and the presence of

polypeptide messengers of inflammation, for instance cytokines and chemokines.

Furthermore, the activation of the complement system generates some of the most

powerful proinflammatory molecules in the body, mainly the three anaphylatoxins C3a,

C4a and C5a. Complement is activated in different tumors, as many studies have found

complement deposition in the membrane of malignant cells and an elevation of

complement activity in the sera of patients with neoplastic diseases (see section 3.1.).

So, activated complement proteins within the tumor microenvironment must be added

to the inflammatory pressure that dictates malignant growth142.

3.5. Complement and lung cancer

The relationship between complement and lung cancer has been known since

some decades ago, when elevated complement levels correlating with tumor size were

found in lung cancer patients143. Afterwards, different studies have reinforced this first

observation, suggesting that, as it happens in other kind of tumors, complement is

activated by lung cancer cells.

As most cancer cells do, lung cancer cells present inhibitory mechanisms to resist

complement dependent cytotoxicity. It has been shown by immunohistochemistry that

NSCLC tumors express on the cell membrane CD59 and CD46 with variable CD55

immunoreactivity112. Several in vitro studies also revealed that lung cancer cell lines are

extremely resistant to CDC, compared to human nasal epithelium primary cell

Page 53: Facultad de Ciencias - CORE

Introduction

24

cultures134,144, although this effect could not be attributed to the inhibitory action of

mCRPs. Lung cancer is not the only cancer type in which blocking of CD46 and CD55

does not make cancer cells more sensitive to complement145, which suggests that there

are other inhibitory mechanisms for controlling the activation of complement on cancer

cells. In particular, the role of soluble factors is taking on major importance after some

interesting findings. For example, Drs Blom and Okroj, in collaboration with our group,

have demonstrated that functional C4BP and factor I, released from NSCLC cell lines,

are able to decreased C3 deposition and complement-dependent lysis124. In addition, our

group has shown that factor H is frequently expressed and secreted by NSCLC.

Released factor H is able to inhibit the activation of complement in vitro and

downregulation of factor H in A549 cells, a NSCLC cell line that expresses factor H,

reduces its growth in vivo122-123. These results suggest that factor H is protecting

efficiently those cells from complement attack.

Due to the shielding role of complement inhibitors, lung tumor cells may resist

complement attack and contribute to the generation of an inflammatory

microenvironment. As indicated in the previous section, this inflammatory

microenvironment may contribute to the malignant development of lung cells.

Page 54: Facultad de Ciencias - CORE

HYPOTHESIS AND OBJECTIVES

Page 55: Facultad de Ciencias - CORE
Page 56: Facultad de Ciencias - CORE

Hypothesis and objetives

27

HYPOTHESIS

The hypothesis of the present work is that tumor cells express on their surface tumor-

associated antigens that have the potential to activate the complement system. At the

same time, tumor cells have developed mechanisms that make them resistant to

complement cytotoxic effects. The subsequent effects of complement activation on

complement-resistant cancer cells play a pivotal role in the development of malignant

features, ending in tumor progression.

OBJECTIVES

1. To study complement activation by the alternative and classical pathways on

non-small cell lung cancer (NSCLC) cells and normal bronchial epithelial cells.

2. To study the expression, at the mRNA and protein levels, of the main

complement inhibitory proteins in NSCLC and normal bronchial epithelial cells.

3. To study the role of complement in a syngeneic mouse model of lung cancer,

with special interest in tumor-associated characteristics of the microenvironment such as

angiogenesis and immunosuppression.

4. To evaluate the tumor-promoting effects of complement on NSCLC cells.

Page 57: Facultad de Ciencias - CORE
Page 58: Facultad de Ciencias - CORE

MATERIALS AND METHODS

Page 59: Facultad de Ciencias - CORE
Page 60: Facultad de Ciencias - CORE

Materials and methods

31

1. MATERIALS

1.1. Animals

The experimental protocols were reviewed and approved by the Institutional

Animal Care and Use Committee of the University of Navarra.

Mice

C57BL wild-type and C3 knockout female mice of 8-12 weeks (The Jackson

Laboratory) were used for the syngeneic experiments with 3LL cells.

Rabbits

Polyclonal antibodies against the A549 cell line were generated in 2-3 kg female

New Zealand White (NZW) rabbits (San Bernando).

1.2. Clinical samples from lung cancer patients

Samples were collected at the Clínica Universidad de Navarra. All protocols were

approved by the Institutional Ethical Committee, and all patients gave informed

consent.

Plasma from 20 NSCLC patients and 20 healthy donors -matched by age and

smoking history- were used. Tumors included both adenocarcinomas and squamous cell

carcinomas in stages III and IV. Samples were stored at -80ºC.

Bronchoalveolar lavage samples from subjects undergoing diagnostic

bronchoscopy were also included in this study. Control samples (n=24) were obtained

from patients with non-malignant lung disease. Tumor samples (n=57) were obtained

from lung cancer patients with different histologies (both SCLC and NSCLC) in stages

III and IV. Samples were stored at -80ºC. The clinicopathological characteristics of this

series are described in a recent publication146.

1.3. Normal human serum

Normal human serum (NHS) from 12 healthy donors was used as a source of

complement factors. Blood was collected in vacuum tubes (BD Vacutainer) and allowed

to clot for 30 minutes at 37ºC. Then tubes were chilled for 15 minutes and centrifuged

at 3345 g. Serum was separated from clot and centrifuged again in order to get a cleaner

Page 61: Facultad de Ciencias - CORE

Materials and methods

32

serum. All the serum samples were pooled together, divided in aliquots and stored at -

80ºC.

To check that the NHS complement activity was within the normal values, the

alternative pathway hemolytic assay (APH50) was performed following the procedure

explained in section 2.1.5.

Heat inactivated normal human serum (HI-NHS) was obtained by heating NHS

during 30 minutes at 56ºC.

1.4. Plasma

Plasma was obtained for factor H purification. Whole blood was collected in

vacuum tubes containing EDTA (BD Vacutainer) and centrifuged for 15 minutes at 4ºC.

Plasma was separated from cells and buffy coat and kept at -20ºC.

1.5. Human non-small cell lung cancer cells

The following human non-small cell lung cancer (NSCLC) cell lines, available at

the American Type Culture Collection (ATCC), were used:

A549: bronchoalveolar carcinoma.

H157: squamous cell carcinoma.

H1299 and H460: large cell carcinoma.

H23 and HTB58: adenocarcinoma.

H727: carcinoid.

1.6. Immortalized normal human epithelial cells

Two types of immortalized normal epithelial cells were used:

- Immortalized HBECs (human bronchial epithelial cells): established by

introducing Cdk4 and hTERT into normal HBECs147. We used two HBEC lines,

from two different donors: HBEC-3KT and HBEC-10KT. Besides, we used HBEC-

3KT cells stably knocked down for p53 (HBEC-3KT p53). The three cell lines were

kindly provided by Dr JD Minna (University of Texas Southwestern Medical

Center, Dallas).

- BEAS-2B cells: established from a normal human bronchial epithelium and

transformed by an SV40 hybrid virus148-149. BEAS-2B are available at ATCC.

Page 62: Facultad de Ciencias - CORE

Materials and methods

33

1.7. Endothelial cells

Primary HUVECs (human umbilical vein endothelial cells) were isolated from

freshly collected umbilical cords (see protocol in section 2.1.1.), or obtained from

ATCC.

1.8. Murine lung cancer cells

The Lewis lung carcinoma cell line (3LL or LLC) was obtained from ATCC. This

cell line was established from the lung of a C57BL mouse bearing a tumor resulting

from an implantation of a primary Lewis lung carcinoma150.

1.9. Hybridoma

The hybridoma cell line OX-23, which produces a monoclonal antibody against

factor H, was obtained from the European Collection of Cell Cultures (ECACC).

1.10. Factor H

Factor H was purified following the method described by Sim et. al151. The final

concentration of factor H was 450 µg/ml in phosphate buffered saline (PBS: 8 mM

Na2HPO4, 2 mM NaH2PO4, 150 mM NaCl, pH 7.4).

1.11. C5aR-1 antagonist

Acetylatedphenylalanine–(ornithine-proline-(D)cyclohexylalanine-tryptophan-

arginine) or AcF(OP(D)ChaWR, a C5aR-1 antagonist, was used in in vitro and in vivo

experiments for the blockade of C5aR-1. The antagonist was used in vitro at 100 ng/ml

and in vivo at a dose of 1 mg per kg body weight. Mice received an injection every 3

days, starting 4 hours before the inoculation of tumor cells. A control peptide, the cyclic

hexapeptide acetylated phenylalanine–(ornithineproline-(D)cyclohexylalanine-alanine-

(D)arginine or AcF(OP(D)ChaA(D)R), was used at the same concentrations. Both

peptides are derived from the C terminal decapeptide sequence of C5a, and were

modified to produce either complete antagonists of the receptor or peptides with no

activity152. Both peptides were synthesized in PeptideSynthetics (Fareham, UK) with a

purity higher than 95%.

Page 63: Facultad de Ciencias - CORE

Materials and methods

34

2. METHODS

2.1. Methods for the generation of experimental material

2.1.1 Cell culture

a. Cell culture conditions

NSCLC and Lewis lung carcinoma cell lines were maintained in RPMI-1640

growth medium (Lonza) with Glutamax, 10% Fetalclone (Thermo), 100 U/ml penicillin

and 100 µg/ml streptomycin (Lonza). Henceforth, this medium will be referred to as

complete medium. For some experiments, cells were kept in RPMI medium without

Fetalclone or antibiotics; this medium will be referred to as basal RPMI.

HBEC-KT cells were maintained in keratinocyte serum free medium

supplemented with human recombinant EGF and bovine pituitary extract (Gibco).

BEAS 2B cells were maintained in DMEM:Ham’s F-12 (1:1) supplemented with

human recombinant EGF, insulin, transferrin, epinephrine, triiodothyronine, retinoic

acid, hydrocortisone, gentamicin sulfate/amphotericin-B and bovine pituitary extract

(media and supplements were from Lonza).

HUVECs were maintained in EBM-2 basal medium containing human

recombinant EGF, VEGF, FGF, R3-IGF-1, hydrocortisone, ascorbic acid, heparin,

gentamicin sulfate/amphotericin-B and 2% fetal bovine serum (all from Lonza);

henceforth, this medium with supplements will be referred to as EGM-2MV. For some

assays, HUVECs were kept overnight in EBM-2 basal medium. HUVECs were either

commercial (ATCC) or obtained from umbilical cords. Human umbilical cords were

obtained from pregnancies and processed within 24 hours. For extracting the vein

endothelial cells, cords were cleaned with 70% ethanol to wipe out the excess of blood

and both ends were cut. Two canules were inserted into the vein of each extreme and

clamped with a thread. Then 20 ml syringes where attached to them. In the first place,

PBS was pushed through the vein to eliminate the blood within it; then, 15 ml of

colagenase at 145 units/ml were pushed and retained inside the vein. The cord was

incubated for 15 minutes at 37ºC for the enzyme to digest the tissue and liberate

endothelial cells. After incubation, the colagenase solution was collected, the remains

were washed with PBS, and gathered together with the first solution. The suspension of

Page 64: Facultad de Ciencias - CORE

Materials and methods

35

cells was spun down and the pellet resuspended in 199 medium (Lonza) with 10% fetal

bovine serum (FBS), 100 U/ml penicillin and 100 µg/ml streptomycin. Cell suspension

was placed in 25 mm3 flasks previously coated with 0.1% gelatin (Sigma), and

incubated overnight at 37°C in a 5% CO2 atmosphere. One day later, medium was

removed and replaced with EGM-2MV.

The mouse hybridoma cell line OX-23 was grown in complete RPMI medium.

For antibody production, hybridoma cells were kept for 10 days in 175 mm2 flasks

containing 25 ml of protein free medium (CD Hybridoma Medium, Invitrogen)

supplemented with 8 mM L-glutamine.

In all cases, cells were grown at 37ºC in a 5% CO2 atmosphere.

Every month and before cell freezing, a biochemical test for mycoplasma

detection (MycoAlert Mycoplasma Detection Kit, Lonza) was performed. The test

consists in the detection of certain mycoplasma enzymes in the medium. Viable

mycoplasma are lysed and the enzymes react with the kit’s substrate catalyzing the

conversion of ADP to ATP, which is measured by luminescence. To perform the test,

conditioned media from 72-96 hour cell cultures were collected and centrifuged at 500 g

for 5 minutes. Afterwards, 100 µl of medium were mixed with 100 µl of MycoAlert

Reagent and 100 µl of MycoAlert Substrate. Luminescence was measured before and

after adding the substrate. If the ratio between the second and the first measure was

higher than one, the cell culture was considered contaminated and discarded.

b. Subcultures

All the cell lines used in this study were adherent to culture flasks, except 3LL

cells which were semi-adherent. Adherent cells were subcultured by removing growth

medium and rinsing them with PBS without Ca2+ or Mg2+. Then, cells were detached

from the flasks by incubation with trypsin/EDTA (Invitrogen) for approximately 5

minutes at 37ºC. Afterwards, trypsin was neutralized with fresh complete medium, cells

were centrifuged at 470 g for 5 minutes, and the pellet was resuspended in fresh

medium. One half or one fourth of the total volume (depending on the cell line) was

mixed with fresh medium and transferred to the culture flask again. The procedure for

the 3LL cells was the same, except that the medium where the cells were grown was

used to neutralize the trypsin. In that way, the non-adherent cells were subcultured

together with the adherent cells.

Page 65: Facultad de Ciencias - CORE

Materials and methods

36

c. Freezing and thawing method

Cells were frozen in order to preserve and maintain a stock of all the cell lines.

For freezing, cells were grown until they reached an 80% confluence. After

trypsinization and neutralization of the trypsin with fresh medium, cells were

resuspended at a concentration of approx. 5 x 106 cells/ml in freezing medium, which

contained 20% dimethylsulphoxide (DMSO, Sigma) in Fetalclone. Samples were

aliquoted in cryotubes (Nalgene) and placed into an isopropanol container, which was

kept at -80ºC for 24 hours. After this time, the cryotubes were transferred to liquid

nitrogen tanks for long-term storage.

For thawing cells, cryotubes were placed for a few minutes in a 37ºC bath, until

the freezing medium got liquid. Then, cells were transferred rapidly to 15 ml tubes with

fresh medium and centrifuged at 470 g for 5 minutes, in order to get rid of the DMSO.

The pellets of cells were seeded in 25 mm3 flasks filled with fresh medium and

incubated at 37ºC in the cell incubator until the confluence was 80-90%. Then, cells

were trypsinized and transferred to 100 mm2 flasks.

2.1.2 Generation of OX-23 anti-factor H antibodies

a. Hybridoma culture

After thawing, the hybridoma cell line OX-23 was grown and subcultured in

complete RPMI medium until confirmation of antibody production by an immunoassay

(see section 2.1.2.e). Once confirmed, cells were grown in protein free medium (CD

Hybridoma Medium, Invitrogen) supplemented with 8 mM glutamine. Conditioned

medium from cells incubated for three days was checked again in order to confirm the

antibody production in the new medium.

b. Production of conditioned media

In order to produce mouse IgG1 monoclonal antibodies against factor H, 20

million OX-23 hybridoma cells were seeded in 175 mm2 flasks with 25 ml of protein

free hybridoma medium supplemented with 8 mM L-glutamine. Cells were grown in

this medium for 10 days, without any change, to increase antibody concentration.

Afterwards, conditioned medium was collected in 50 ml tubes and centrifuged for 5

minutes at 470 g. Cells were discarded, and supernatants from different flasks were put

Page 66: Facultad de Ciencias - CORE

Materials and methods

37

together and kept at -20ºC until antibody purification. An aliquot was saved apart to test

its immunoreactivity (see section 2.1.2.e).

c. Purification

For antibody purification, 100 ml of supernatant was used. First, proteins were

precipitated by mixing the supernatant with 76.1 g of ammonium sulfate (Sigma), added

carefully to obtain a saturated dilution. The solution was maintained with slight shaking

during 16 hours at 4ºC. Then, the solution was filtrated using a Whatman paper and a

Buchner funnel connected to a vacuum pump. The precipitate was recovered from the

paper by placing it in a 50 ml tube with 10 ml of PBS and shaking it slightly for five

minutes. Then, the paper was discarded and the solution was centrifuged for 5 minutes

at 3345 g at 4ºC. The paper-free solution was introduced into a dialysis membrane and

placed on a 500 ml beaker filled with PBS. The dialysis was performed at 4ºC for 2

days. The buffer was changed twice. After dialysis, the immunoreactivity was evaluated

in an aliquot of the solution (see section 2.1.2.e). Monoclonal antibodies within the

solution were purified by chromatography (AKTApurifier, Amersham) using a HiTrap

Protein G column (Amersham). The elution buffer was 100 mM glycine (Sigma) at pH

2.5. The elution buffer was neutralized by adding 50 µl of 1 M Trizma-base (Sigma) at

pH 9. Finally, the antibodies were dialyzed against PBS as mentioned above.

d. Determination of protein concentration

Protein concentration was measured with the Bradford reagent (BioRad). A BSA

(bovine serum albumin) standard curve was used. Samples or standards (10 µl) were

added to the plates in duplicate; then, 190 µl of Bradford reagent diluted 1:5 in distilled

water was added to the wells. After 10 minutes of incubation, absorbance at 595 nm and

450 nm was measured in a Sunrise microplate reader with the Magellan software

(Tecan).

e. Titration

An immunoreactivity assay was performed in order to evaluate the presence of

OX-23 antibodies in four different samples: conditioned medium previous to the

purification procedure, solutions obtained previous and after HPLC purification, and

final dialyzed solution.

Page 67: Facultad de Ciencias - CORE

Materials and methods

38

For the assay, 50 ng of purified factor H (Sigma) in PBS was added per well in

one half of a polystyrene 96 well plate (flat bottom, non-sterile, Costar). The other half

was filled with 50 µl of PBS per well, and was used to calculate the background for

each antibody dilution. Factor H was allowed to attach to the plate for 1 hour at room

temperature. Then, wells were washed three times with assay buffer (PBS with 1% BSA

and 0.1% Tween-20; all reagents from Sigma) and blocked with 200 µl of the same

buffer overnight at 4ºC. Afterwards, 50 µl of serial dilutions of the samples in assay

buffer (from 1:10 to 1:106) was added in triplicate onto wells with and without factor H.

As a positive control, a previous OX-23 batch was used in the same conditions. Plates

were incubated at room temperature for 2 hours; then, wells were washed twice with

assay buffer, and 50 µl of peroxidase-conjugated rabbit anti-mouse IgG antibody

(Amersham) diluted 1:1000 in assay buffer was added, and incubated at room

temperature for 30 minutes. After two washes, the assay was developed with 200 µl of

Sigmafast OPD (o-phenylenediamine dihydrochloride), previously prepared according

to the manufacturer’s instructions. Absorbance was measured at 450 nm in a microplate

spectrophotometer (Tecan).

f. SDS-PAGE and Coomassie staining

To check antibody purity, an electrophoresis was performed in a polyacrylamide

gel with sodium dodecyl sulfate (SDS) under reducing and non-reducing conditions,

followed by Coomassie staining (Figure 5).

Precast 4-12% Bis-tris gradient gels (NuPAGE, Invitrogen) were used. Ten

microliters of purified antibody (5 µg of proteins, approximately) were mixed with

commercial loading buffer NuPAGE LDS Sample Buffer (Invitrogen). For reducing

conditions, 10% β-mercaptoethanol was also added. The mix was boiled for 5 minutes,

snap-cooled to avoid protein refolding, and loaded into the gel.

Electrophoresis was carried out during 1 hour at 120 V in running buffer (50 mM

MOPS, 50 mM Trizma-base, 0.1% SDS and 1 mM EDTA, pH 7.7; all from Sigma).

After electrophoresis, gels were carefully placed in cuvettes, and rinse with ultrapure

water. Then, cuvettes were filled with a Coomassie blue solution (Pierce) and gels were

incubated with slight shaking for one hour. To remove excess of colorant, the gels were

washed three times with ultrapure water and photographed in a digital gel

documentation system (Gel Doc, BioRad).

Page 68: Facultad de Ciencias - CORE

Materials and methods

39

150 kDa

45 kDa

30 kDa

150 kDa

45 kDa

30

OX 24 OX 23

RNRNR R

150 kDa

45 kDa

30 kDa

150 kDa

45 kDa

30

OX 24 OX 23

RNRNR R

Figure 5. Coomassie staining obtained after electrophoresis in reducing (R) and non reducing conditions (NR) of purified OX23 antibody. OX24 antibody, previously purified in our laboratory, was used as positive control. Staining shows three bands: in non reducing conditions the 150 kDa band corresponds to the complete antibody molecule; and in reducing conditions the 45 kDa and 30 kDa bands correspond to the heavy and the light chains of the antibody, respectively.

2.1.3 Anti-A549 polyclonal antibody generation

a. Production of antibodies in NZW rabbits

Before the procedure, A549 cells were grown and tested for mycoplasma. Cells

were cultured in 175 mm2 flasks until 80% confluence, and then were harvested. Three

aliquots with 107 cells in 500 µl of PBS were prepared and kept at -80ºC until

inoculation. Female NZW rabbits were also prepared before the procedure as follows.

The hair from the dorsal area was removed using hair-removing cream (Veet), and

periphery blood was collected by venepuncture of the marginal ear vein in order to

obtain preimmune serum. For this purpose, blood was allowed to clot at 4ºC for 16

hours and then centrifuged at 677 g on a benchtop centrifuge for 15 minutes. Serum was

transferred to new tubes and centrifuged again. Finally, serum was collected, heated for

30 minutes at 56ºC to inactivate complement, and kept at -20ºC until further use.

Immunization was performed by three injections of 107 cells at 15-day intervals.

The first injection was prepared by mixing cells with 500 µl of Complete Freud

Adjuvant (Difco Laboratories) using the Sonifier 150 (Branson) for 30 seconds. The

emulsion formed was transferred to a 1 ml syringe (BD Plastipack) and maintained on

ice till the moment of injection. Then, it was injected intradermically in six different

spots on the dorsal area of the rabbit, using a 25G needle (BD Microlance). Fifteen days

later, another emulsion was prepared with 107 cells, but in this case with 500 µl of

Incomplete Freud Adjuvant (Difco Laboratories). The injection was done this time

Page 69: Facultad de Ciencias - CORE

Materials and methods

40

subcutaneally in two different spots of the dorsal area of the rabbit. Another fifteen days

later, the third and last emulsion was prepared in the same way as the second one, but

was injected intramuscularly in one spot on a footpad of the rabbit.

Fifteen days later from the last injection, whole blood from rabbits was collected.

For the procedure, animals were anesthetized with 35 mg/kg ketamine (Imalgene 1000,

Merial), 5 mg/ml xylazine (Rompun 2%, Bayer) and 1 mg/ml acepromazine (Calmo

Neosan; Tpizer). Blood was collected directly from the heart using 10 ml syringes with

25G needles. After transferring it to 50 ml tubes, blood was allowed to clot and then

centrifuged. The antiserum was finally prepared by heating the supernatant for 30

minutes at 56ºC. Then, it was aliquoted and maintained at -20ºC for further

experiments.

b. Immunoreactivity assay

Immunoreactivity against A549 cells was confirmed by flow cytometry. A549

cells (105) were incubated with 50 µl of serial dilutions of the antiserum in binding

buffer (PBS with 1% BSA and 0.1% sodium azide; all reagents from Sigma), ranging

from 1:25 to 1:200; or preimmune serum diluted 1:25 as a negative control. After 30

minutes at 4ºC, cells were washed twice with binding buffer and resuspended in 50 µl of

FITC conjugated anti-rabbit IgG antibody (Molecular Probes) diluted 1:100 in binding

buffer. Cells were again incubated for 30 minutes at 4ºC and washed twice in binding

buffer. Finally, cells were resuspended in 100 µl of FACS Flow (BD Bioscience) with

7-amino-actinomycin D (7AAD; Sigma) at 50 µg/ml and incubated for 10 minutes at

4ºC protected from light. Afterwards, cells were analyzed in a FACSCalibur flow

cytometer with the CellQuest Pro software (BD Biosciences).

The same assay was later performed in all NSCLC and normal epithelium cell

lines, to check the ability of the antibody to bind to the different cell types.

2.1.4 Factor H purification

To purify factor H from plasma, we followed a method already described151.

a. Preparation of columns

Purification of factor H and FHL1 implicates passing of plasma through three

different columns previously prepared according to manufacturer’s instructions.

Page 70: Facultad de Ciencias - CORE

Materials and methods

41

The first one, a lysine-Sepharose column (Lysine Sepharose TM 4B; Amersham),

removes the plasmin/plasminogen. The second one, the preadsorption column, is a

column of IgG-conjugated Sepharose (AminoLink Plus Immobilization Kit), that

removes proteins that bind to immunoglobulins or to modified agarose (e.g. C1q,

fibronectin, rheumatoid factors). This column was prepared in three phases, which

implies: protein immobilization, blockade of remaining active sites, and column

washing; according to the manufacturer’s instructions. The third column contains the

monoclonal antibody anti-factor H OX-24 immobilized on Sepharose, and retains factor

H and FHL-1.

b. Factor H purification

For factor H purification, columns were equilibrated in running buffer (PBS with

15 mM EDTA, pH 7.2). Plasma was prepared by adding EDTA (Sigma) and Pefabloc

[4-(2-aminoethyl) benzenesulfonyl fluoride hydrochloride, AEBSF, Fluka], both to a

final concentration of 0.5 mM. Plasma (5 ml) was added to the first column, where the

plasmin/plasminogen was retained, and eluted with running buffer. Pefabloc was added

again to the elute to made a final concentration of 0.5 mM. The sample was added to the

second column, and eluted with running buffer. Finally, the elute was passed through

the third column, where factor H was retained. The column was then washed with TBS

(25 mM Tris-HCl, 140 mM NaCl, 0.5 mM EDTA, pH 7.4) and factor H and FHL-1

were eluted with 3M MgCl2.

The elute was then dialyzed twice; first, against TBS for 16 hours at 4ºC, and

second, against PBS for 24 hours. The final solution was concentrated by centrifugation

at 2000 g and 4ºC using Amicon centricons with a 10 kDa cutoff (Millipore).

c. Factor H integrity

To check factor H purity and integrity, an electrophoresis (SDS-PAGE) and

Coomassie staining were done as explained above (section 2.1.2.f). Under reducing

conditions, the gel showed two bands of 150 and 42 kDa, which correspond to FH and

FHL-1, respectively (Figure 6).

Page 71: Facultad de Ciencias - CORE

Materials and methods

42

150 KDa

c FH p FH

NR R

c FH p FH

42 KDa

150 KDa

c FH p FH

NR R

c FH p FH

42 KDa

Figure 6. Factor H and FHL-1 purification. Coomassie staining obtained after electrophoresis in reducing (R) and non reducing conditions (NR) of purified factor H and FHL-1 (p FH). Comercial factor H (c FH) was used as positive control. Staining showed two bands corresponding to factor H and FHL-1.

d. Factor H quantification by ELISA

An ELISA assay was performed in order to calculate factor H concentration at

different stages throughout the procedure. For this purpose, several aliquots were

collected from these solutions: starting plasma, elutes from each column, elute in the

concentration step, and final solution.

To measure factor H/FHL-1 concentration, a polystyrene 96-well plate (flat

bottom, non sterile, Costar) was coated with 50 ng/well of anti-factor H monoclonal

antibody OX-23 in 50 µl of 50 mM sodium bicarbonate (pH 8), and incubated for 1

hour at room temperature. After three washings with assay buffer (PBS with 1% BSA

and 0.1% Tween 20; all from Sigma), plates were blocked overnight at 4ºC with the

same buffer. Afterwards, 50 µl of each sample collected throughout the purification

procedure, or serial dilutions of commercial factor H (Sigma) ranging from 1.5 to 200

ng/ml, were added in triplicate, and the plate was incubated for two hours at room

temperature. Wells were washed three times with assay buffer and 50 µl of goat anti-

factor H antibody (Quidel) diluted 1:1000 in assay buffer was added. After 30 minutes

of incubation at room temperature, wells were washed three times and incubated for 30

minutes with peroxidase-conjugated polyclonal rabbit anti-goat IgG antibody (Dako)

diluted 1:4000 in assay buffer. Finally, the assay was developed with OPD (Sigma),

according to manufacturer’s instructions. The plate was read at 450 nm in a microplate

reader (Tecan). The final concentration of factor H was 450 µg/ml.

Page 72: Facultad de Ciencias - CORE

Materials and methods

43

2.1.5 Alternative pathway hemolytic assay (APH50)

APH50 was performed as previously described153. This method is used to

determine the amount of complement required to lyse 50% of red cells in 1 ml of a

solution that contains 108 erythrocytes/ml. It is expressed in APH50 units and in humans

its value is comprised between 38 and 93 U/ml154.

Rabbit erythrocytes were used for the procedure, as they activate the alternative

pathway of complement spontaneously. For this purpose, blood from female NZW

rabbits was collected directly from hearts as explained in section 2.1.3. After allowing

blood to clot for 2 hours, the pellet of cells was transferred to a 50 ml tube and washed

three times with ten times the pellet’s volume in Alsever’s solution (113.7 mM

dextrose, 27.2 mM sodium citrate, 2.6 mM citric acid, 72 mM sodium chloride, pH 6.2;

all reagents from Sigma) at 677 g during 10 minutes, and resuspended in this solution

for maintenance at 4ºC.

For the procedure, a 108 erythrocytes/ml dilution is needed. To obtain this

concentration of cells, 1 ml of cells kept on Alsever’s solution was centrifuged, and the

pellet resuspended in 15 ml of GVB+ (3.1 mM barbituric acid, 1.8 mM barbital, 145

mM NaCl, 0.1 % gelatin, 0.1 M EGTA, 0.1 mM MgCl2, pH 7.4). Erythrocytes in 100 µl

of this solution were lysed with 2900 µl of distilled water. Then, serial dilutions were

prepared (1:2, 1:4 and 1:8) and their absorbances measured at 412 nm. Beer-Lambert

equation was used to calculate each dilution’s concentration knowing that 0.294 is the

absorbance of a 108 erythrocytes/ml dilution. Afterwards, a solution with 2 x 108

erythrocytes/ml was prepared in GVB+ using the stock solution maintained in Alsever’s.

Hemolytic assays were performed in U bottom 96-well plates (Costar). Different

volumes: 50, 40, 30, 20, 10 or 5 µl, of 1:2 diluted serum in GVB+ were added in

triplicate. A final volume of 50 µl was obtained adding GVB+. For the 0% and 100%

lysis, 50 µl of GVB+ or distilled water were added, respectively. Then, 50 µl of 2 x 108

erythrocyte/ml solution was added to each well and the plate incubated for 30 minutes

at 37ºC with slight agitation. Afterwards, 150 µl of chilled 0.9% NaCl (Sigma) was

added to each well and the plate centrifuged at 1000 g for 5 minutes at 4ºC.

Supernatants (120 µl) were transferred to flat bottom 96-well plates, and absorbance

was read at 412 nm in a microplate reader (Tecan).

Page 73: Facultad de Ciencias - CORE

Materials and methods

44

To determine APH50, first, the percentage of lysis (y) for each serum dilution was

calculated as:

Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

y =Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

y = x 100Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

y =Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

y =Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

y =Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

Absorbance serum dilution x – Absobance 0% lysis

Absobance 100% lyisis – Absorbance 0% lysis

y = x 100

Then, the von Krog equation for the sigmoid curve of hemolysis was used:

x = K (y/(1-y))1/n

In this case, x is the volume of serum added to the tube (µl) and 1/n is 0.2 ± 10%,

depending on conditions used. K is a constant. At the point of 50% hemolysis:

(y/(1-y))1/n = 1, hence x = K

To calculate the value of K, first, the value of (y/(1-y)) for each serum dilution was

calculated, and then, log x was plotted against log (y/(1-y)) (Figure 7). The intercept on

the x-axis was K´ (as log 1= 0). K was the K´ antilog.

The number of APH50 U/ml of the original serum was obtained as:

APH50 = (1000/K) x 2 (which is the dilution of serum in our particular case).

-20

0

20

40

60

80

100

120

5 10 20 30 40 50-1,2

-0,8

-0,4

0

0,4

0,8

1,0 1,3 1,5

Lysis

(%

)

Lo

g (

y/(

1-y

))

Serum volume (µl) Serum volume (Log)

A B

-20

0

20

40

60

80

100

120

5 10 20 30 40 50-1,2

-0,8

-0,4

0

0,4

0,8

1,0 1,3 1,5

Lysis

(%

)

Lo

g (

y/(

1-y

))

Serum volume (µl) Serum volume (Log)

A B

Figure 7. Calculating APH50. (A) The graph shows the percentage of lysis vs. volume of diluted serum for a test sample. (B) The graph shows a log-log plot of (y/(1-y)) vs. volume of diluted serum. The interception with X-axis corresponds to K’ value. In this particular example K value was 22.5 µl of serum, which corresponded to 88.8 U/ml of APH50.

In our experiments we used pools of sera with APH50 values within the normal

values in humans, 38 and 93 U/ml154.

2.1.6 Total RNA extraction and retrotranscription

To extract total RNA from cell cultures, cells were seeded in 100 mm2 plates until

reaching 80% confluence. Then, cell medium was removed and 1 ml of Ultraspec

(Biotecx) was added. Cell homogenates were detached from plates using a scraper,

mixed with 200 µl of chloroform and vortexed. Mixtures were laid for 5 minutes on ice

Page 74: Facultad de Ciencias - CORE

Materials and methods

45

and then centrifuged at 12,000 g at 4ºC for 15 minutes. The aqueous phase was

carefully transferred to a clean tube and the same volume of isopropanol was added.

Then, tubes were slightly mixed and kept for 10 minutes at 4ºC or frozen at -80ºC, as

cold helps precipitation. After the precipitation step, samples were centrifuged for 10

minutes at 12,000 g and 4ºC. The white pellets were washed twice with 50 µl of 75%

ethanol in DEPC water (Sigma) and dried at 50ºC. Finally, extracted RNA was

resuspended in 20 µl DEPC water.

From frozen tumors, RNA was extracted using the RNeasy Micro kit (QIAGEN).

Before starting the protocol, samples were homogenized by mixing the tissues in a hand

held homogenizer with three-four crystal beads, a little spoon of quartz (Merk) and

700 µl of β-mercaptoethanol diluted 1:100 in RLT buffer (provided in the kit). After

three cycles of shaking, the mixtures were centrifuged at high speed for 3 minutes and

supernatants were transferred into QIAshredder spin columns (QIAGEN). The columns

were centrifuged at high speed for 3 minutes in a benchtop centrifuge and the elutes

were used for RNA extraction following the RNeasy Micro kit protocol.

RNA samples extracted from cultured cells or tumor tissues were analyzed for

integrity by agarose gel electrophoresis, and their concentrations measured using a

Nanodrop 1000 spectrophotometer (Thermo Scientific). RNA was stored at -80ºC until

further use.

Complementary DNA (cDNA) was obtained by standard retrotranscription, using

the SuperScript III kit (Invitrogen).

For the procedure, the following mixture was prepared:

Total RNA (extracted from cells or tissues) 1 µg

Random deoxynucleotide hexamers (60 ng/µl) 1 µl

dNTPs (2.5 mM, each) 1 µl

DEPC water (Sigma) up to 11 µl

The mixture was incubated for 5 minutes at 65ºC in a PTC-100 thermal cycler

(BioRad). After this first step of denaturalization, samples were snap-cooled on ice, to

avoid formation of secondary structures within the RNA template, while the following

mixture was prepared:

Page 75: Facultad de Ciencias - CORE

Materials and methods

46

First strand reaction buffer (5X) 4 µl

DTT (0.1 M) 1 µl

RNase OUT (40 U/µl) 1 µl

SuperScript III 1 µl

This mixture was added to samples and the retrotranscription was performed in

the thermal cycler using two temperature steps: 50 minutes at 50ºC for the enzyme

reaction, and 15 minutes at 70ºC for the enzyme denaturalization. Then, samples were

placed on ice and 1 µl of RNase H (5 U/µl, Invitrogen) was added in order to remove

RNA templates. Tubes were warmed for 20 minutes at 37ºC and, finally, cDNA was

stored at -20ºC.

2.1.7 Protein extraction

For protein extraction, cells were washed twice with PBS and then lysed during

15 minutes at 4ºC in RIPA buffer (10 mM Tris, 150 mM NaCl, 1% Triton X-100, 1%

sodium deoxycholate and 0.1% sodium dodecyl sulfate, pH 8; all from Sigma) with one

tablet per 10 ml of protease inhibitor cocktail (Roche). Samples were centrifuged at high

speed on a benchtop centrifuge and supernatants containing dissolved proteins were

transferred to clean tubes.

Protein concentration was measured with the Bradford reagent (BioRad) as

explained in section 2.1.2.d.

2.1.8 Genotyping of C3 deficient mice

Genotyping of C3 deficient mice was performed every six months as follows. Ear

biopsies from C3 knockout mice and wild type controls were digested at 55°C overnight

in lysis buffer (1 M Tris, 0.5 M EDTA, 10% SDS, 5 M NaCl and 0.1 mg/ml proteinase

K, pH 8.5). After centrifugation at high speed for 5 minutes to remove debris,

supernatants were transferred to new tubes and the same volume of 100% isopropanol

(Panreac) was added to them, in order to precipitate genomic DNA. After precipitation,

the pellet was washed in 70% ethanol, dried at room temperature for 10 minutes, and

resuspended in ultrapure water. Finally, DNA concentration was measured using the

Nanodrop 1000 spectrophotometer (Thermo Scientific). Genotyping was performed by

conventional PCR following the protocol and primers from The Jackson Laboratory

(Table 3).

Page 76: Facultad de Ciencias - CORE

Materials and methods

47

Table 3. Primers used in the genotyping of wild-type and C3 knockout mice by conventional PCR.

2.2. Methods for the study of complement activity on cells

2.2.1 Complement deposition on cells after activation of the alternative or

classical pathways

For the activation of the alternative pathway of complement, cells were detached

from culture dishes with trypsin/EDTA (Lonza), washed twice with PBS, and

resuspended in veronal buffer with magnesium (1.8 mM barbital, 3.1 mM barbituric

acid, 141 mM NaCl, 0.5 mM MgCl2, pH 7.4; hereafter referred as VB+). Then, cells (2 x

105) were incubated with 10% NHS or HI-NHS for 30 minutes at 37ºC. After two

washes in PBS, deposition of C3, iC3b, C5b or C5b-9 was assessed by incubating cells

with the specific primary antibodies diluted in binding buffer (PBS with 1% BSA and

0.1% sodium azide; all from Sigma) as follows: 1:100 FITC-conjugated polyclonal goat

anti-human C3 (ICN Biomedicals), 1:200 monoclonal mouse anti-human iC3b

neoantigen (Quidel), 1:100 monoclonal mouse anti-human C5b-9 (clone aE11; Dako),

or 1:100 polyclonal goat anti-human C5 (Quidel). After 30 minutes at 4ºC, cells were

washed twice in binding buffer and incubated in 1:100 Alexa Fluor-conjugated goat

anti-mouse IgG (Invitrogen) or FITC-conjugated rabbit anti-goat IgG (Invitrogen) for

30 minutes at 4ºC. Afterwards, cells were washed twice in binding buffer and

resuspended in 100 µl of FACS flow (BD Biosciences) with 7-AAD at a final

concentration of 50 µg/ml. After ten minutes of incubation with the dye, flow

cytometric analyses were performed.

For the classical pathway activation, cells were treated as explained above but

resuspended in veronal buffer with magnesium and calcium (1.8 mM barbital, 3.1 mM

barbituric acid, 141 mM NaCl, 0.5 mM MgCl2, 0.15 mM CaCl2, pH 7.4; hereafter

referred as VB2+). Cells (2 x 105) were incubated with the rabbit antiserum against A549

cells diluted 1:50 in a total volume of 100 µl. After two washes with VB2+, cells were

resuspended in 10% NHS or HI-NHS in VB2+. From this step forward, cells were

treated as explained for the alternative pathway.

Gene Sense Antisense

C3 WT 5´ATCTTGAGTGCACCAAGCC3´ 5´GGTTGCAGCAGTCTATGAAGG3´

C3 MUT 5´CTTGGGTGGAGAGGCTATTC3´ 5´AGGTGAGATGACAGGAGATC3´

GAPDH 5´ACTTTGTCAAGCTCATTTCC3´ 5´TGCAGCGAACTTTATTGATG3´

Page 77: Facultad de Ciencias - CORE

Materials and methods

48

2.2.2 Complement cytotoxicity after activation of the alternative or classical

pathways

The lactate dehydrogenase (LDH) assay, which measures membrane integrity as a

function of the amount of cytoplasmic LDH released into the medium, was used to

determine cytotoxicity due to complement activation on the cell membrane. LDH

activity is determined in a coupled enzymatic reaction: in the first step, NAD+ is

reduced to NADH/H+ by the LDH-catalyzed conversion of lactate to pyruvate. In the

second step, the catalyst (diaphorase) transfers H/H+ from NADH/H+ to the

iodotetrazolium chloride (tetrazolium salt INT) which is reduced to formazan, that is

detected by spectrophotometry.

LDH assay (Cytotoxicity detection kitPLUS, Roche) was performed according to

the manufacturer’s instructions, using the supernatants from cells treated with

complement. In order to obtain the supernatants, 10,000 cells were prepared in GVB2+

(1.8 mM barbital, 3.1 mM barbituric acid, 141 mM NaCl, 0.5 mM MgCl2, 0.15 mM

CaCl2, and 0.1% gelatin, pH 7.4), or in GVB+ (GVB2+ without CaCl2) for the activation

of the classical and alternative pathways, respectively. In the case of the classical

pathway, cells were preincubated for 30 minutes at 4ºC with the antiserum against A549

cells diluted 1:50 in GVB+, and washed twice in the same buffer before incubation with

the human serum. Cells were incubated in GVB+ or GVB2+ containing 10% NHS, 10%

HI-NHS (background lysis control) or 0.5% Triton X-100 (100% lysis control) for 30

minutes at 37ºC; all conditions were prepared in triplicate. After complement activation,

cells were pelleted in a benchtop centrifuge at 500 g for 5 minutes at 4ºC. Supernatants

were recollected for the LDH test.

For the assay, 50 µl of supernatants diluted 1:5 in PBS were placed in triplicate in

a 96-well plate, that made 9 repetitions per condition. In order to subtract the

background absorbance of the serum and Triton X-100, 50 µl of 2% NHS, 2% HI-NHS

and 1% Triton X-100 were added in triplicate. Then, 50 µl of freshly prepared reaction

mixture (one part of Catalyst, that contains diaphorase/NAD+ mixture, per 45 parts of

Dye solution, which contains INT and lactate) were added in each well and incubated

25 minutes at room temperature. The reaction was stopped by adding 25 µl of Stop

solution, that contains Triton X-100 and hydrochloric acid. Finally, absorbance was

measured at 492 nm with a reference wavelength of 690 nm in a microplate reader

(Tecan).

Page 78: Facultad de Ciencias - CORE

Materials and methods

49

Percentage of lysis was determined using the mean value of triplicates, by the

following formula:

(AbsorbanceNHS – Absobancebackground NHS)-(AbsorbanceHI-NHS – Absobancebackground HI-NHS)

(Absobance100% lyisis - Absorbancebackground 100% lysis)Lysis (%) = x 100

(AbsorbanceNHS – Absobancebackground NHS)-(AbsorbanceHI-NHS – Absobancebackground HI-NHS)

(Absobance100% lyisis - Absorbancebackground 100% lysis)

(AbsorbanceNHS – Absobancebackground NHS)-(AbsorbanceHI-NHS – Absobancebackground HI-NHS)

(Absobance100% lyisis - Absorbancebackground 100% lysis)Lysis (%) = x 100

2.2.3 C5a quantification in plasma samples and bronchoalveolar lavages by

ELISA

C5a concentration in plasma samples from patients with NSCLC or healty donors,

and in BALs from patients with lung cancer (SCLC or NSCLC) or with non-malignant

lung disease, were quantified using the commercial kit Human Complement Component

C5a (R&D), following the manufacturer’s instructions. Prior to the analysis, samples

were spun down at 300 g for 10 min, and the supernatants were collected.

2.2.4 Study of complement inhibitors in NSCLC and normal epithelial cells

a. Determination of mRNA levels by quantitative PCR

Real time PCR was performed to analyze the expression of the main complement

inhibitors on NSCLC and normal bronchial epithelial cells. Total RNA was isolated

from cell cultures and reverse transcribed as described in section 2.1.6.

For the PCR reaction, a mix containing the following components was prepared

per well, all samples were done in triplicate:

cDNA 0.2 µl

Sybr Green PCR Master Mix (Applied Biosystems) 12.5 µl

Primers S and AS (10 µM each), listed in Table 4 0.75 µl each

Ultrapure water up to 25 µl

Reactions were performed using the 7300 Real Time PCR System (Applied

Biosystems) according to the following scheme:

50°C for 2 minutes

95°C for 10 minutes

95°C for 15 seconds

60°C for 1 minute

And an extra dissociation stage, which consists on a continuous increase of

temperature while measuring fluorescence. This permits the detection of the

40 cycles

Page 79: Facultad de Ciencias - CORE

Materials and methods

50

temperature at which drastic decrease of fluorescence takes place, due to double strand

uncoupling and Sybr Green dye detaching from DNA. This temperature is the amplicon

Tm.

Relative levels of expression were assessed by the threshold cycle (Ct) values and

expressed as a percentage relative to the glyceraldehyde 3-phosphate dehydrogenase

(GAPDH) mRNA.

Table 4. List of primers used for the analysis of complement inhibitors by quantitative PCR.

*FH primers were designed to recognize the 17th exon; therefore, they recognize factor H but not FHL1; ** FH/FHL-1 primers were designed to recognize the second exon of the gen; therefore, they recognize both factor H and FHL-1.

b. Factor H binding to the cell membrane

Binding of factor H/FHL-1 was assessed in NSCLC and normal epithelial cells by

incubating 105 cells in binding buffer (PBS with 1% BSA and 0.1% sodium azide; all

reagents from Sigma) with purified factor H (450 µg/ml in PBS; see section 2.1.4.)

diluted 1:4 in binding buffer or 25% HI-NHS. After 30 minutes of incubation at 37ºC,

cells were washed twice in binding buffer and incubated with goat anti-factor H primary

antibody (Quidel) diluted 1:100 in binding buffer for 30 minutes at 4ºC. Afterwards,

cells were washed in binding buffer and incubated with FITC-conjugated rabbit anti-

goat IgG (Molecular Probes), diluted 1:100 in binding buffer. Finally, cells were

washed twice and incubated with 50 µg/ml of 7-AAD for 10 minutes in the dark, then,

analyzed by flow cytometry in a FACScalibur with CellQuest Pro software (BD

Biosciences).

c. Detection of membrane-bound complement inhibitors

Expression of CD55, CD59 and the protein-kinase casein kinase 2 (CK2) was

assessed in both NSCLC and normal bronchial epithelial cells. Cells were detached

Gene Sense Antisense

FH* 5´GATGTGTATAAGGCGGGTGAG3´ 5´GGAGGTGTCTCTGCATGTTG3´

FH/FHL-1** 5´GAAGGCACCCAGGCTATCTA3´ 5´ATCTCCAGGATGTCCACAGG3´

FI 5´GAGGAAAGCGAGCACAACTG3´ 5´GTCGGGGTGTATCCAGTCTACTA3´

CD46 5´GTGCTGCTCCAGAGTGTAAAGTG3´ 5´CGCTGCCATCGAGGTAAA3´

CD55 5´CCACAAAAACCACCACACC3´ 5´GCCCAGATAGAAGACGGGTAGTA3´

CD59 5´GGAATCCAAGGAGGGTCTGT3´ 5´CAGTCAGCAGTTGGGTTAGGA3´

GAPDH 5´GAAGGTGAAGGTCGGAGTC3´ 5´GAAGATGGTGATGGGATTTC3´

Page 80: Facultad de Ciencias - CORE

Materials and methods

51

from the culture dishes with trypsin/EDTA (Lonza), washed twice with PBS, and

resuspended in binding buffer. Cells (2 x 105) were washed twice with binding buffer

and incubated for 30 minutes at 4ºC with mouse anti-human CD55 (BRIC 216,

IBGRL), rat anti-human CD59 (YTH51, Serotec), both diluted 1:25 in binding buffer,

or mouse anti-human CK2 (1AD9, Sigma) diluted 1:100 in binding buffer. After

incubation, cells were washed twice in binding buffer and incubated for 30 minutes in

the dark with Alexa Fluor-conjugated goat anti-mouse IgG (Invitrogen) or FITC-

conjugated rabbit anti-rat IgG (Serotec), both at 1:100 dilution in binding buffer.

Finally, inhibitor expression was analyzed by flow cytometry in a FACScalibur with the

CellQuest Pro software (BD Biosciences).

d. Elimination of GPI-anchored proteins from the cell membrane by PI-PLC

treatment

In some experiments, CD55 and CD59 were removed from NSCLC and normal

epithelial cells by a treatment with the enzyme phosphatidylinositol-specific

phospholipase C (PI-PLC) from Bacillus cereus (Molecular Probes). PI-PLC cleaves the

phospholipid phosphatidylinositol (PI), which is part of the molecule

glycosylphosphatidylinositol (GPI) that anchors hundreds of proteins, including CD55

and CD59, to the cell membrane.

To remove these complement regulators from the cell membrane, cells (2 x 105)

were incubated one hour at 37ºC in 100 µl of basal RPMI with 0.5% BSA and one unit

of PI-PLC. Afterwards, cells were washed twice in PBS. These cells were used to

determine C5b-9 deposition and cell cytotoxicity due to the activation of the alternative

pathway of complement.

e. Effect of hypoxic conditions in the expression of complement inhibitors in

NSCLC cells

In order to mimic hypoxic conditions, cells were incubated at 37°C in a

humidified hypoxia chamber (Hypoxia Workstation 400; Ruskinn Technology),

connected to a Ruskinn gas mixer module supplying 94% N2, 5% CO2 and 1% O2. Cells

were either kept in the hypoxic chamber for 48 hours (referred to as hypoxia) or for 24

hours followed by 24 h at normal O2 tension (referred to as hypoxia/reoxygenation).

Control cells (normoxia) were cultured for 48 hours under normal O2 tension. Cells

Page 81: Facultad de Ciencias - CORE

Materials and methods

52

were then used to analyze the expression of complement inhibitors at both the mRNA

and protein levels.

2.3. Methods to study the in vitro effects of C5a

2.3.1 C5aR-1 expression

a. C5aR-1 mRNA expression

Conventional PCR was used for the screening of C5aR-1 mRNA expression.

Total RNA was extracted from cells and retrotranscribed to cDNA. The PCR reaction

mixture was as follows:

Master Mix buffer (Promega) 12.5 µl

Primer S and AS (10 µM each), listed in Table 5 1 µl

cDNA 2 µl

Ultrapure water up to 25 µl

The reactions were performed according to the following conditions:

94ºC 5 minutes

94ºC 45 seconds

56º 45 seconds

72ºC 1 minute

72ºC 5 minutes

To identify PCR products, electrophoresis in agarose gels was performed. Gels

were casted by mixing agarose (Pronadisa) 1% w/v with 1x Tris-acetate-EDTA buffer

(TAE), then, the mixture was boiled in order to disolve the agarose. Ethydium bromide

(Invitrogen) was added at a final concentration of 0.5 µg/ml. Gels were left to solidify

into gel casters (BioRad). Prior to electrophoresis, samples were mixed with loading

buffer (0.25% bromophenol blue, 0.25% xylenecyanol and 50% glycerol; all from

Sigma) in a 1:5 proportion. Samples were loaded and at least one well per gel was

preserved for a 100 bp molecular weight marker. Electrophoresis was performed at

100 V during 45 minutes. Afterwards, imaging and analysis of the gels was done in a

digital gel scanner (Gel Doc, BioRad).

30 cycles

Page 82: Facultad de Ciencias - CORE

Materials and methods

53

Table 5. List of primers used in the analysis of C5aR-1 by conventional PCR.

b. C5aR-1 protein expression

Cells (2x105) were incubated for 30 minutes at 4ºC with a mouse anti-human

C5aR-1 (S5/1, Serotec) or an IgG2b isotype control (R&D Systems) diluted 1:50 or

1:100, respectively, in binding buffer (PBS with 1% BSA and 0.1% sodium azide; all

reagents from Sigma). After two washes, cells were incubated in Alexa Fluor-

conjugated goat anti-mouse IgG (Invitrogen) diluted 1:100 in binding buffer for 30

minutes at 4ºC. Finally, cells were washed twice and analyzed by flow cytometry on a

FACSCalibur using CellQuest Pro software (BD Biosciences).

2.3.2 Treatment with C5a

The following treatment with C5a was performed for most experiments; however,

C5a concentrations and incubation times may differ, as it will be indicated in the

corresponding section.

Treatment of cells with C5a (Calbiochem) was performed in 10 cm2 plates. Cells

(5 x 105) were seeded in complete medium and left to attach for 24 hours at 37ºC in the

cell incubator. Afterwards, cells were washed three times with 10 ml of PBS and kept in

basal RPMI overnight. Next day, the medium was removed and 10 ml of fresh basal

medium (negative control) or basal medium containing C5a was added. In some

experiments, the C5aR-1 antagonist was used to study the specificity of C5a; in those

cases an additional plate was treated with C5a (10 ng/ml) and C5aR-1 antagonist (100

ng/ml). Cells were incubated for different periods of time. In some cases, conditioned

media were also collected at the end of the experiments and concentrated using Amicon

centricons with a 10 kDa cutoff (Millipore).

2.3.3 C5a signaling activation

a. Calcium influx assay

Granulocytes were used as positive controls. They were obtained from fresh

heparinized blood by centrifugation at 750 g during 10 minutes. Then, the pellet was

resuspended in lysis buffer (55 mM NH4Cl, 10 mM KHCO3, pH 7.6) and incubated for

Gene Sense Antisense

C5aR-1 5´ATGAACTCCTTCAATTATACC3´ 5´TGGTGGAAAGTACTCCTCCCG3´

GAPDH 5´ACTTTGTCAAGCTCATTTCC3´ 5´CACAGGGTACTTTATTGATG3´

Page 83: Facultad de Ciencias - CORE

Materials and methods

54

5 minutes at room temperature for erythrocytes elimination. After incubation, the

dispersion of cells was centrifuged and the pellet washed with basal RPMI. Cells were

counted and prepared at 8x106 cells in 500 µl of basal RPMI. Tumor cells were left 24

hours in basal medium before the experiment, and then trypsinized and resuspended

(8x106) in 500 µl of basal RPMI. To each sample, 1 µl of Fluo-4 (Millipore) was added,

and incubated for 30 minutes at 37ºC in the dark. Cells were then washed twice and

resuspended in 500 µl of basal RPMI. The increase in fluorescence due to changes in

the intracellular calcium content was measured on the FACScalibur flow cytometer

after gating the proper population, granulocytes (in the case of whole blood) or tumor

cells. Baseline fluorescence was established by measuring Fluo-4 labeled cells during 2

minutes. After that, either 10 µl of C5a at 100 µg/ml or PBS were added, and changes in

calcium mobilization were measured for an additional 3 minutes. Data were analyzed

using the FlowJo software (Tree Star).

b. ERK phosphorylation

A549 cells were treated for 45 minutes with or without C5a, as explained in

section 2.3.2. Proteins extracted in RIPA buffer (30 µg) were mixed with commercial

loading sample buffer (NuPAGE LDS Sample Buffer; Invitrogen) and 1 µl of β-

mercaptoethanol. The mixture was boiled during 10 minutes at 70ºC. Afterwards,

samples were snap-cooled to avoid protein refolding, and loaded into the gel. To

estimate the molecular weight of proteins under study, a standard commercial molecular

weight marker, MultiMark (Invitrogen), was used. Samples were loaded into

commercial denaturing gels of 4-12% polyacrilamide (NuPAGE Bis-Tris; Invitrogen)

and the electrophoresis was carried out in an Xcell SureLock system (Novex) in running

buffer (50 mM MOPS, 50 mM Tris, 0.1% sodium dodecyl sulfate and 1 mM EDTA, pH

7.7; all from Sigma), during 45 minutes at 180 V. After electrophoresis, proteins were

transferred to 0.45 µm nitrocellulose membranes (BioRad) in an Xcell II Blot Module

system (Invitrogen) during 90 minutes at 30 V. Afterwards, membranes were rinsed

twice with PBS-0.1%Tween (PBST), and blocked overnight with blocking solution

(PBST with 10% skim milk) at 4ºC. Next day, blots were incubated for one hour with a

rabbit anti-human phospho p44/p42 MAPK (Thr202/tyr204) and a rabbit anti-human

p44/p42 MAPK (both from Cell Signaling) diluted 1:2000 in blocking solution. After

three washes with PBST (10 minutes each), blots were incubated for 30 minutes at room

temperature with a horseradish peroxidase-conjugated anti-rabbit IgG secondary

Page 84: Facultad de Ciencias - CORE

Materials and methods

55

antibody (Amersham) diluted 1:2000 in blocking solution. Afterwards, membranes were

washed three times with PBST and immunoreactive bands were visualized by

chemoluminiscence using the Lumi-LightPLUS kit (Roche) following the manufacturer’s

instructions. Blots were slightly dried and placed into radiography cassettes together

with ECL Amersham Hyperfilm sheets during variable amounts of time, and

radiography sheets were developed automatically in a Curix 60 device (AGFA).

c. NF-κκκκB translocation to the nucleus

NF-κB translocation to the nucleus was tested in A549 cells treated with C5a.

Cells (10,000) were cultured in 8-well glass chambers (BD Biosciences) in complete

RPMI and incubated for 48 hours at 37ºC and 5% CO2. Cells were then washed twice

and left in basal medium overnight. Next day, cells were treated in basal medium with

10 ng/ml C5a, with or without 100 ng/ml C5aR-1 antagonist or control peptide. Basal

medium was used also as negative control. After 45-minute incubation, cells were

washed twice with PBS and fixed in 4% paraformaldehyde for 15 minutes at room

temperature. After two additional washes with PBS, cells were permeabilized with 0.5%

Tween in PBS for 10 minutes, blocked with 10% goat serum in PBS and incubated

overnight at 4ºC with mouse 1:200 anti-NF-κB antibody (Santa Cruz Biotech) in PBS

containing 1% BSA. Cells were washed with PBS and incubated with 1:200

phycoerythrin (PE) anti-rabbit IgG secondary antibody (Invitrogen) for 1 hour at room

temperature. Finally, cells were stained with 10 µl of DAPI and visualized by using an

Axio Imager ZI with Axio Vision software (Zeiss).

2.3.4 Effect of C5a on cell proliferation

A colorimetric MTT assay, which is a means of measuring the activity of living

cells by assessing the activity of mitochondrial dehydrogenases, was used to study the

effect of C5a on cell proliferation. Mitochondrial dehydrogenase activity on viable cells

is measured by a reduction reaction, where yellow MTT [3-(4,5-dimethylthiazol-2-yl)-

2,5-diphenyltetrazolium bromide] is reduced to formazan crystals, giving a purple color.

The building up of crystals inside the cells causes their lysis and release of cellular

content to the exterior. Then formazan crystals, which are insoluble in aqueous

solutions, are dissolved in a solubilization solution that contains DMSO.

The assay was used to study the proliferation of HUVEC and NSCLC cell lines.

For endothelial cells, 1500 HUVECs were seeded in 50 µl of assay medium (EGM-

Page 85: Facultad de Ciencias - CORE

Materials and methods

56

2MV medium without rhVEGF and rhFGF) in a 96-well sterile plate (Costar). After 24

hours of incubation, 50 µl of C5a (Calbiochem) or bFGF (Sigma) were added in assay

medium two times more concentrated than the desired final concentrations. bFGF at a

final concentration of 10 ng/ml was used as positive control, and cells with only assay

medium were used to assess 100% proliferation. All treatments were performed in six

wells. In order to subtract the background absorbance of the medium, 100 µl of medium

was added in six empty wells. Cells were placed in the cell incubator for 96 hours.

Afterwards, 10 µl of 5 mg/ml MTT solution (Sigma) was added to each well. Plates

were incubated for an additional 4 hours at 37ºC to allow MTT reduction by viable

enzymes. Afterwards, 100 µl of solubilization buffer (10% SDS, 50% N-N-

dimethylformamide; both from Sigma) was added and plates were left on the cell

incubator overnight. Next day, absorbance was measured at 540 and 690 nm

(corresponding to formazan and reference wavelengths, respectively) in a Sunrise

microplate reader (Tecan). In some experiments, bFGF was also added to the assay

medium at a final concentration of 0.31 ng/ml, in order to induce cell proliferation.

For NSCLC cell lines, the procedure was similar to that explained for HUVEC. In

this case, 1500 NSCLC cells (A549, H157 and H661 cells) were seeded in 50 µl of

RPMI complemented with 1% Fetalclone and allowed to attach overnight. Next day,

50 µl of C5a two times more concentrated than the final concentration was added in

basal RPMI (this made a medium with 0.1% Fetalclone). In some experiments, medium

was removed before the addition of C5a and cells were washed twice in basal medium.

Then, C5a dilutions in basal medium were added to cells; so no serum was present.

Cells were incubated for 96 hours at 37ºC. The MTT reaction and development was

done as indicated above for HUVEC.

To measure the percentage of proliferation, the mean value of each condition was

calculated and used in the formula:

Absorbance treatment concentration – Absobance background

Absobance – Absorbance background

Absorbance treatment concentration – Absobance background

Absobance 100% proliferation – Absorbance background

Proliferation (%) = x 100Absorbance treatment concentration – Absobance background

Absobance – Absorbance background

Absorbance treatment concentration – Absobance background

Absobance 100% proliferation – Absorbance background

Proliferation (%) = x 100

2.3.5 Effect of C5a on cell cycle

A549 cells were treated with C5a as indicated in section 2.3.2. Afterwards, cells

were harvested with trypsin/EDTA and washed twice in PBS. For fixation, cells (1x106)

were resuspended in 5 ml of 70% cold ethanol and left at 4ºC for at least one hour (this

step can be extended for one week). After fixation, cells were washed twice in PBS and

Page 86: Facultad de Ciencias - CORE

Materials and methods

57

the pellets resuspended in 500 µl of RNase (Invitrogen) at 200 µM in PBS. Then, cells

were incubated for one hour at 37ºC and, finally, 20 µl of propidium iodide at 1 mg/ml

was added. Cells were analyzed in the FACScalibur flow cytometer.

2.3.6 Effect of C5a on anchorage independent growth

A549 cells were treated with C5a as indicated in section 2.3.2. Anchorage-

independent assays were performed by covering 6-well plates with 1.5 ml of 0.6%

sterile noble agar (BD Biosciences) dissolved in 1% medium (RPMI with 1%

Fetalclone, 100 U/ml penicillin and 100 µg/ml streptomicin). Cells (1000 cells/well)

were mixed with 1 ml of 0.3% agar dissolved in 1% medium with or without 10 ng/ml

C5a. Plates were incubated for 8, 10 and 12 days at 37ºC. After incubation, 500 µl of 10

mg/ml MTT was added to each well and plates were incubated again at 37ºC for

additional 4 hours. Finally, excess of MTT was removed carefully with a pipete, 500 µl

of DMSO was added to wells, and plates were incubated at 37ºC overnight. For

analyzing the number of colonies in each well, plates were digitally scanned and

colonies were counted.

2.3.7 Effect of C5a on cell migration

The effect of C5a on cell migration was evaluated in HUVEC and A549 cells

using a transwell migration assay. This assay consists in seeding cells in specially

designed inserts with microscopic pores in 24-well plates (Corning-Costar). Then, a

chemoattractant solution is added where the inserts are laid. If cells are attracted to the

chemoattractant, they will migrate through the pores of the transwells and keep attached

to the lower side of them (in case they are attaching cells).

HUVEC (75,000) and A549 (10,000) cells were seeded on the upper chamber of

transwells in 150 µl of EBM-2 or RPMI basal medium, respectively. In both cases,

chambers with 8 µm pore size filters were used. C5a dilutions ranging from 500 to

0.25 ng/ml in EBM-2 or RPMI basal medium were added in the 24-well plates, in a

final volume of 400 µl. EBM-2 or RPMI basal medium was used as negative control,

and 10% FBS in EBM-2 or RPMI basal medium as positive control. Cells were allowed

to migrate for 48 hours at 37ºC with 5% CO2. After incubation, cells from the upper

chambers were removed by swiping with cotton swabs, and transwells were washed

twice in PBS. Cells adhered to the lower side of transwells were fixated with 4%

formaldehyde (Panreac) for one hour at room temperature. Afterwards, cells were

Page 87: Facultad de Ciencias - CORE

Materials and methods

58

stained with 0.5% crystal-violet (Sigma) on PBS for 15 minutes at room temperature.

Excess of staining was removed by washing twice with PBS. Migrated cells on the

lower surface of the filter were visualized using an inverted microscope (Leyca DM

ILM), and at least five fields at 10x magnification were photographed (Leica EC3

digital camera). Migrated cells were then counted using ImageJ software (NIH).

2.3.8 Effect of C5a on HUVEC tube formation

C5a ability to promote angiogenesis in vitro was tested with the tube formation

assay using HUVECs. For the assay, HUVECs were used between the 5th and 7th

passages and were cultured in 10 cm2 plates coated with 0.1% gelatin (Sigma). Cells

were cultured in EGM-2MV until they got an optimal confluence (between 80-90%);

then, the medium was changed to EGM-2MV/EBM-2 basal medium (1:1) and cells

were incubated for 8 hours. The reason to use this transition medium (with two-fold

diluted supplements) is to avoid a sudden change from the normal conditions to a total

lack of supplements. Afterwards, cells were washed and kept in EBM-2 basal medium

overnight. Next day, 48-well plates were coated with 150 µl of growth factor reduced

Matrigel (BD Biosciences) and the matrix was allowed to gel for 2 hours at 37ºC.

HUVECs (25,000) were seeded in the gelled Matrigel in 150 µl of EBM-2 basal

medium, and 150 µl of 20 ng/ml C5a in EBM-2 basal medium, or only 150 µl of EBM-

2 basal medium (negative control), was added to cells. At least five repetitions were

done per condition and plates were incubated at 37ºC for 5 hours. Afterwards, the

medium was carefully removed by turning the plates upside down, and the cells were

fixed and stained with Diff Quik (Panoptico Rapido; QCA) following the

manufacturer’s instructions, and handling the plate with care, as tubes are very fragile

structures. Photographs of three random optical fields at 10x magnifications in the

central area of wells were taken with a digital camera coupled to a phase microscope

(Leica DM IL LED). Total length of tubes in each well was calculated using the ImageJ

software (NIH).

2.3.9 Effect of C5a on cell adhesion to extracellular matrix proteins

This assay was performed in order to study the effect of C5a on the adhesion of

NSCLC cells to several intracellular matrix components, as previously described by

Irigoyen et al.155

Page 88: Facultad de Ciencias - CORE

Materials and methods

59

NSCLC cells, A549, H157 and H661, were treated as indicated in section 2.3.2.

For the adhesion assay, 96-well plates with flat bottom (Costar) were cover with 35 µl

of different matrix dilutions: 5 µg/cm2 rat tail collagen type I (BD Bioscience) in

20 mM acetic acid, 5 µg/cm2 collagen type IV (Sigma) in 50 mM acetic acid, 1 µg/cm2

fibronectin (Sigma) in PBS, 0.1 µg/cm2 vitronectin (Sigma) in ultrapure water, 1 µg/cm2

laminin (Sigma) in PBS, or 0.1 µg/cm2 gelatin (Sigma) in ultrapure water; 3% BSA in

PBS was used as a negative control. All matrix dilutions were added in sextuplicate.

Plates were incubated for 2 h at 37°C. After incubation, they were washed twice with

PBS, and blocked with 200 µl of 1% BSA in PBS for 1 h at 37°C. At the same time,

cells were trypsinized, washed twice in PBS, and incubated in 300 µl of basal RPMI

containing 10 µM calcein-AM (Fluka) for 20 min at 37°C, in a water bath. After

labeling, cells were washed three times in basal RPMI to eliminate excess of calcein-

AM, and pellets were resuspended in a concentration of 2 × 105 cells/ml in adhesion-

medium (RPMI with 0.5% BSA and 20 mM HEPES). Before adding cells to the plates,

the blocking solution was removed from wells. Then, 100 µl of each cell dilution

(control cells, cells treated with C5a, and cells treated with C5a and C5aR1 antagonist)

was added in the different matrixes. The remaining cells were maintained for the last

step (see below). Plates were incubated for 30 minutes at 37ºC in order to allow cells to

attach to the matrixes. After the incubation time, non-adherent cells were removed by

gentle washing with adhesion-medium; the remains of medium were wiped carefully

with a multi-channel pipette. Finally, 100 µl of basal RPMI were added to the attached

cells and, to establish the 100% adhesion, 100 µl of each cell dilution (that were kept

previously for this step) were added in six empty wells. Fluorescent adherent cells were

measured on a Polar Star Galaxy plate reader (BMG Labtechnologies), with an

excitation wavelength of 485 nm and an emission filter of 520 nm.

To calculate the percentages of adhesion to the different matrixes in each

condition, fluorescence mean of replicates was calculated, and the following formula

was used:

Fluorescence cells adhered to matrix – Fluorescence cells adhered to 3% BSA

Fluorescence 100% adhesion – Fluorescence cells adhered to 3% BSA

Adhesion (%) = x 100 Fluorescence cells adhered to matrix – Fluorescence cells adhered to 3% BSA

Fluorescence 100% adhesion – Fluorescence cells adhered to 3% BSA

Fluorescence cells adhered to matrix – Fluorescence cells adhered to 3% BSA

Fluorescence 100% adhesion – Fluorescence cells adhered to 3% BSA

Adhesion (%) = x 100

2.3.10 Effect of C5a on integrin expression

Integrin expression on the cell membrane was analyzed by flow cytometry. Cells

were treated as explained in section 2.3.2. for 48 hours with 10 ng/ml C5a. Cells (2x105)

Page 89: Facultad de Ciencias - CORE

Materials and methods

60

were incubated with primary monoclonal antibodies against integrins α1, α4, αv, β1,

β3, α5β1, αvβ3, and vβ4. All antibodies came from the Integrin Classics Kit

(Chemicon International) and were diluted 1:100 in binding buffer (PBS with 1% BSA

and 0.1% sodium azide; all reagents from Sigma), except for the antibody anti-αvβ4,

which was from BD Pharmigen and was diluted 1:40. An isotype control anti-IgG2b

(R&D Systems) diluted 1:10 was also used. After 30 minutes of incubation, cells were

washed twice in binding buffer and incubated for 30 minutes in Alexa fluor goat anti-

mouse IgG (Invitrogen) diluted 1:100 in binding buffer. Then, after two washes in

binding buffer, cells were analyzed by flow cytometry in a FACScalibur with the

CellQuest software (BD Biosciences).

2.3.11 Effect of C5a on MMP activity

For measuring the matrix metalloproteinase (MMP) activity on the conditioned

media, fluorogenic peptide substrates were used, those peptides incorporate a

fluorophore and a quencher moiety positioned on either side of the enzyme cleavage

site. When an active protease cleaves the substrate, the fluorescence is increased

(Figure 8). The increase in fluorescence correlates with the amount of protease activity.

Donor

QuencherRecognition

site

Enzyme

Generation of

fluorescent signal

Donor

QuencherRecognition

site

Enzyme

Generation of

fluorescent signal Figure 8. Schematic representation of the fluorogenic peptide recognition by specific proteases and subsequent cleveage and fluorescence generation. (Adapted from LABTECH).

For the assay, conditioned media from cells treated as explained in section 2.3.2.

with 10 ng/ml C5a, with or without 100 ng/ml C5aR-1 antagonist, were collected after

24 or 48 hours and concentrated using Amicon centricons with a 10 kDa cutoff

(Millipore). Concentrated media were incubated in triplicate with the fluorogenic

peptides Mca-R-P-K-P-V-E-Nval-W-R-K(Dnp)-NH2 and Mca-PLGL-Dpa-AR-NH2

(R&D Systems), at a final concentration of 20 µM, in TCNB buffer (50 mM Tris, 150

Page 90: Facultad de Ciencias - CORE

Materials and methods

61

mM NaCl, 10 mM CaCl2 and 0.5% Brij, pH 7.5). As a positive control, MMP-10 at 20

nM was used; and as an inhibition control, 100 µM GM 6001 (Chemicon International)

was added to the samples with the highest metalloproteinase activity. GM 6001 is a

wide range hydroxamate MMP inhibitor. Plates were incubated at 37ºC for 2 hours

while the increase of the fluorescence intensity during substrate cleavage was monitored

in a fluorescence spectrofluorimeter (SpectraMAX GeminiXS, Molecular Devices). The

measurements were carried out at the excitation λex=320 nm and at the emission

λem=405 nm. The increase in fluorescence was plotted against the incubation time and

the slope (rate of increase in fluorescence) was calculated by linear regression.

2.4. Mice models for the study of the influence of complement activation on lung

tumor growth in vivo

2.4.1 Syngeneic animal model of lung cancer

The murine Lewis Lung Cancer cell line (also called LLC or 3LL) implanted in

the flanks of female C57BL/6J mice, a model of syngeneic lung cancer, was used to

analyze the effect of complement in tumor progression, angiogenesis and immune

response against tumors.

For the experiments, 3LL cells were trypsinized and neutralized with the

conditioned medium, as this cell line is a semi-adherent cell culture, and then washed

twice with PBS. Mice were anesthetized with a mixture containing 35 mg/kg ketamine

(Imalgene 1000, Merial) and 5 mg/ml xylazine (Rompun 2%, Bayer). Two different

inoculation sites were used: subcutaneous in the mouse back, and intravenous in the tail

vein. For subcutaneous tumor growth, 2.5x104 cells were resuspended in 100 µl of PBS

and mixed with 100 µl of growth factor reduced Matrigel (BD Biosciences). Two

hundred microliters of the cell/Matrigel mixture was injected subcutaneously into the

flanks of mice (wild type or C3-deficient mice). Diameters of the tumors were measured

every three days with calipers, and the volume was calculated by the formula:

(L × W2)/2, where L is the length and W is the width. Around the 30th day after cell

injection, animals were sacrificed and spleen and tumors were removed, weighed, and

processed for flow cytometry analysis or used for total RNA extraction, as explained in

section 2.4.2. For intravenous injection, mice received an injection into the tail vein of

1x105 cells in 100 µl of PBS. After 3 weeks, mice were anesthetized as above and

Page 91: Facultad de Ciencias - CORE

Materials and methods

62

sacrificed by exsanguination. Lungs were harvested and fixed as explained in section

2.4.2.d.

In some experiments, C5aR-1 was blocked pharmacologically in wild type

C57BL mice to study specifically the effect of C5a through its main receptor. Mice

received an injection every 3 days of C5aR-1 antagonist [AcF(OP(D)ChaWR)] or

control peptide [AcF(OP(D)ChaA(D)R)], at a dose of 1 mg per kg body weight in

100 µl of PBS, starting 4 hours before the inoculation of tumor cells.

In some experiments, C3 deficient animals were treated with cobra venom factor

(CVF, Aczon) in order to eliminate any possible effect of C5 in those animals. CVF

forms a stable C3/C5 convertase with factor B; this characteristic allows it to cleave

both complement components C3 and C5, resulting in complement depletion. In a pilot

experiment, CVF was injected in wild type mice in order to set the administration

timing by monitoring C3 levels in serum. From the pilot experiment a dose of 5 µg per

gram body weight in 100 µl of PBS injected intraperitoneally was settled, with the

following regimen of injections: three injections at 28, 24 and 4 hours before cell

inoculation (in order to deplete complement activity before cell implantation), and

injections every 3 days after inoculation (to avoid complement recovery during the

experiment). Complement depletion was monitored by ELISA quantification of C3 in

mouse sera. This assay was also used to confirm C3 deficiency in C3 knockout mice.

C3 ELISA was carried out in 96-well plates coated with goat anti-mouse C3 (Cappel

Laboratories), diluted 1:1000 in 50 mM sodium bicarbonate, pH 8.3. After 1 hour at

room temperature, plates were washed in the assay buffer (PBS with 1% BSA and 0.1%

Tween, all reagents from Sigma) and blocked with the same buffer. Afterwards, 1:1000,

1:2000 and 1:4000 dilutions of samples in assay buffer were added in triplicate. When

the ELISA was used to confirm C3 deficiency in C3 knockout mice, dilutions of a pool

of sera from 8 normal wild type mice ranging from 1:500 to 1:256,000 were also added

in triplicate. Plates were placed at room temperature for 2 hours and then washed twice

with assay buffer. Finally, the assay was developed using a horseradish peroxidase-

conjugated goat anti-mouse C3 antibody (Cappel) diluted 1:5000 in assay buffer, and

OPD (Sigma), according to manufacturer’s instructions. Plates were read at 450 nm in a

Sunrise microplate reader (Tecan). Percentage of C3 depletion in mice treated with CVF

was calculated by setting the C3 levels before CVF injection as 100%. C5a levels were

also measured in wild type and C3 knockout mice. For this purpose the mouse

Page 92: Facultad de Ciencias - CORE

Materials and methods

63

complement component C5a DuoSet (R&D Systems) was used following the

manufacturer’s instructions.

2.4.2 Tissue processing

a. Cell isolation from spleens

Spleens were weight and cut in two parts. To obtain single-cell suspensions, one

portion was mechanically disaggregated in 5 ml of PBS using cell strainers with 70 µm

pore size (BD Biosciences). Cells were then centrifuged at 500 g during 5 minutes and

resuspended in 500 µl of PBS. Single-cell suspensions were used to calculate total cell

numbers in spleens, using a Neubauer chamber, and to analyze, by flow cytometry, the

main populations within the spleens.

Single-cell suspensions from spleens (approx. 5 x 105 splenocytes) were

centrifuged and incubated with primary antibodies. Antibodies were diluted in binding

buffer (PBS with 1% BSA and 0.1% sodium azide; all reagents from Sigma) following

the conditions summarized in Table 6. In the case of Treg, they were fixated,

permeabilizated and stained according to the manufacturer’s instructions. After 30

minutes of incubation in each antibody dilution, splenocytes were washed twice and

analyzed by flow cytometry on the FACSCalibur with CellQuest Pro software (BD

Biosciences).

Table 6. Antibodies used for staining splenocytes*

*All antibodies, except those for Tregs, were from BD Pharmingen. Antibodies for Tregs were obtained on the kit for mouse regulatory T cells from eBioscience. MDSC: myeloid-derived suppressor cells.

Cell population Fluorochrome-conjugated monoclonal antibody Dilution

T cells CD4+ FITC-conjugated rat anti mouse CD4 (RM4-5) 1:1000

T cells CD8+ PE-conjugated rat anti mouse CD8a (53-6.7) 1:1000

B cells CD19+ FITC-conjugated rat anti mouse CD19 (ID3) 1:400

MDSC FITC-conjugated rat anti mouse CD11b (M1/70) 1:500

APC-conjugated rat anti mouse Ly6C/Gr1 (AL-2) 1:100

Tregs FITC-conjugated rat anti mouse CD4 (RM4-5) 0.125 µg/test (1:40)

APC-conjugated rat anti mouse CD25 (PC61.5) 0.06 µg/test (1:30)

PE-conjugated rat anti mouse Foxp3 (FJK-16S) 0.2 µg/test (1:20)

Page 93: Facultad de Ciencias - CORE

Materials and methods

64

b. Total RNA extraction from tumors and quantitative PCR

Total RNA extracted from tumors was analyzed for the expression of different

molecules related to the immune response and angiogenesis. Portions from the edge of

tumors with a volume of approx. 0.1 cm3 (a sphere with 0.3 cm radio) were cut, frozen

in dry ice, and maintained at -80ºC until the extraction procedure. Total RNA was

extracted and retrotranscribed to cDNA as explained in section 2.1.6. Quantitative PCR

was performed by Drs Lasarte and Riezu, from the Division of Gene Therapy &

Hepatology at CIMA. RNA expression levels of the different genes in the study were

related to β-actin expression levels.

c. Fixation of tumors

Once the portion for RNA extraction was obtained, tumors were fixed in 4%

formaldehyde for 24 hours.

d. Fixation of lungs

Lungs harvested from mice receiving an injection into the tail vein of 3LL cells

were fixed by intratracheal instillation of 4% formaldehyde at a constant pressure of 20

cm H20156. One day later, lungs were transferred to 70% ethanol and paraffin blocks

were made from each lobe in the Morphology Core Facility at CIMA. One section per

block was cut and stained with hematoxylin and eosin, resulting on a total of five slides

per mouse. Lung nodules were counted in a microscope. Consecutive slices to those

with nodules were used for CD31 immunohistochemistry.

2.4.3 Study of angiogenesis by CD31 immunohistochemistry

Slices of fixed lungs from the intravenous injection model of lung cancer were

used. Fixed lung sections were kept at 60ºC during 20 minutes to allow paraffin

melting. Immediately, they were placed in xylol (Panreac) for 15 minutes to remove

paraffin leftovers. Then, sections were hydrated with decreasing alcohol stages for 5

minutes: 96%, 80%, 70% ethanol, and finally rinsed in distilled water for another 5

minutes. To block endogenous peroxidases, sections were incubated with 3% hydrogen

peroxide (Merck) for 10 minutes. Afterwards, they were rinsed and subjected to antigen

unmasking. This step consisted in boiling the sections in citrate buffer (10 mM citrate,

pH 6.0) in a standard microwave oven for a maximum of 10 minutes in two consecutive

times. After that, slides were kept at room temperature for 5 minutes to cool down, and

Page 94: Facultad de Ciencias - CORE

Materials and methods

65

washed in TBS (50 mM Tris, 150 mM NaCl, pH 7.4). An incubation of one hour with

1:20 anti-mouse CD31 (LabVision) in REAL antibody diluent (Dako) was done. Slides

were washed in TBS for five minutes and incubated with 1:50 rabbit anti-rat IgG

(Dako) during 30 minutes. After another washing step, samples were incubated in

Envision anti-rabbit (Dako) for 30 minutes. Samples were revealed by incubation with

diaminobenzidine for 8 minutes. The reaction was stopped with water. Finally, samples

were stained with hematoxylin and dehydrated with increasing alcohol solutions to be

finally driven to xylol and mounted with DPX (VWR). Images were capture using a

microscope with a digital camera and ACT-2U software (Nikon). Quantification of the

microvascular density on tumor sections was carried out by measuring the fraction of

area immunostained using the AnalySIS FIVE software (Olympus BioSystems).

2.5. Statistical analysis

The effect of hypoxic conditions in the expression of mCRPs and soluble factors

was evaluated by ANOVA and the Tukey post-test. The Mann-Whitney test was used

for evaluation of the expression of different citokines within the tumors, and for

evaluation of the differences of factor H in bronchoalveolar lavage supernatants. The

other comparisons were carried out with the Student’s t test. Relationship between

variables was quantified using Spearman’s rank correlation. Box plots were used to

present the results of factor H and C5a quantification on clinical samples. In these plots,

the bottom and top of the boxes represent the lower and upper quartiles, the band inside

the box is the median, and the whiskers the minimum and maximum of all the data.

Receiver operating characteristic (ROC) curves were used to define the best cut-off

point and evaluate the performance of the factor H test. Area under the ROC curve, as

well as the 95% confidence interval, was also calculated. Data were analyzed using

SPSS software or GraphPad Prism (GradPad Software). For all analyses, p values of

0.05 or less were considered statistically significant.

Page 95: Facultad de Ciencias - CORE
Page 96: Facultad de Ciencias - CORE

RESULTS

Page 97: Facultad de Ciencias - CORE
Page 98: Facultad de Ciencias - CORE

Results

69

1. COMPLEMENT ACTIVATION AND REGULATION ARE

INCREASED IN NON-SMALL CELL LUNG CANCER CELLS

Initially, complement activation and its regulation were compared in vitro between

non-small cell lung cancer (NSCLC) cells and normal bronchial epithelial cells. In

terms of activation, both classical and alternative pathways were studied, and deposition

of complement proteins on the cell membrane and cell cytotoxicity were analyzed. With

respect to regulation, expression of the main complement inhibitors was analyzed, as

well as the effect of hypoxia on the expression of these inhibitors.

1.1. Deposition of complement proteins after activation of the alternative

pathway

Seven human NSCLC cells lines were chosen for these experiments, representing

the principal subgroups of the disease. The activation of the alternative complement

pathway in these cell lines was compared with that in four immortalized human normal

bronchial epithelial cells.

Activation of the alternative pathway of complement was analyzed by measuring

the increase of C3 deposition on the cell membrane in cells treated with 10% NHS

versus cells treated with 10% HI-NHS (Figure 9A). A significant increase in C3

deposition was exhibited in all NSCLC cell lines, except HTB58, compared to BEAS-

2B normal bronchial epithelial cells (pA549=0.002; pH23<0.001; pH157<0.001;

pH460=0.030; p H727=0.002; pH460=0.011). No significant differences were observed

between the different normal bronchial epithelial cells analyzed. This result indicates

that there is an increase in spontaneous complement activation on malignant lung cells

when compared to normal lung epithelial cells. In order to determine if there was any

difference in C3b inactivation among cells, detection of iC3b on the surface of cells was

carried out (Figure 9B). In each cell line, the amount of iC3b on the membrane was

comparable with the amount of C3b, and the significant differences with normal

epithelial cells were maintained (pA549= 0.007; pH23=0.017; pH157=0.010; pH460=0.038;

pH727<0.001; pH1299=0.029; pHTB58=0.003). Finally, we analyzed the deposition of C5b

on the cell membrane (Figure 9C). Compared to C3b and iC3b deposition, this factor

Page 99: Facultad de Ciencias - CORE

Results

70

appeared at lower levels on the surface of all the cell lines tested, but the differences

between groups were still significant (pA549=0.005; pH23=0.044; pH157=0.025;

pH727=0.006; pH1299=0.040; pHTB58<0.001).

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

10

20

30

C3 d

epositio

n (M

FI ra

tio)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

20

40

60

80

iC3b d

epositio

n (M

FI ra

tio)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

BEA

S

0

1

2

3

4

C5 d

epositio

n (M

FI ra

tio)

∗∗

∗∗∗

∗∗∗

∗∗

∗∗∗

∗∗

∗∗∗

∗∗∗

∗∗

∗∗∗∗

∗∗∗

A B

C

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

10

20

30

C3 d

epositio

n (M

FI ra

tio)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

20

40

60

80

iC3b d

epositio

n (M

FI ra

tio)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

BEA

S

0

1

2

3

4

C5 d

epositio

n (M

FI ra

tio)

∗∗

∗∗∗

∗∗∗

∗∗

∗∗∗

∗∗

∗∗∗

∗∗∗

∗∗

∗∗∗∗

∗∗∗

A B

C

Figure 9. Deposition of complement proteins on NSCLC (shaded bars) and normal bronchial epithelial cells (white bars) after activation of the alternative complement pathway. Graphs show flow cytometry data of cells incubated in VB+ with 10% NHS vs. HI-NHS (as a negative control), and stained for C3b (A), iC3b (B) and C5b (C). At least three independent experiments for each cell line were performed. Results are expressed as the ratio of the mean fluorescence intensity (MFI) between NHS and HI-NHS, and shown as mean ± SEM. Statistical differences were analyzed using BEAS cells as reference. *p<0.05; **p<0.01; ***p<0.001.

1.2. Deposition of complement proteins after activation of the classical pathway

In order to activate the classical pathway, two antibodies were evaluated, an

antiserum obtained from rabbits immunized against whole cell extracts of A549 cells and

an antiserum against β2-microglobulin. Both polyclonal antibodies showed high and

similar levels of binding to NSCLC and normal bronchial epithelial cell lines, however

the antiserum against β2-microglobulin failed to trigger the classical complement

pathway (data not shown). Therefore, we used the antiserum against A549 cells to

activate the classical complement pathway on all the cell lines studied (Figure 10).

Page 100: Facultad de Ciencias - CORE

Results

71

Fluorescence intensity

HTB58H1299 HBEC 3KT

HBEC 10KT BEAS

H727

Cell

nu

mb

er

HBEC 3KTp53

H23 H157A549 H460

Fluorescence intensity

HTB58HTB58H1299H1299 HBEC 3KTHBEC 3KT

HBEC 10KTHBEC 10KT BEASBEAS

H727H727H727

Cell

nu

mb

er

HBEC 3KTp53HBEC 3KTp53

H23H23H23 H157H157H157A549 A549 A549 H460

Figure 10. Binding of the polyclonal antibody against A549 cells to NSCLC and normal bronchial epithelial cells. Cells were incubated with serum obtained from rabbits immunized against A549 cell extracts (white peak) or with pre-immune serum (shaded peak), with a FITC-conjugated anti-rabbit IgG, and analyzed by flow cytometry. Binding of the polyclonal antibody to the membrane was measured as the increase in intensity in the green channel.

Complement activation by the classical pathway was analyzed in the same manner

as that used for the alternative pathway. First, C3 deposition was compared between

NSCLC and normal epithelial cells. This pathway is more powerful than the alternative

pathway, so all cell lines showed much higher C3 levels on their cell membrane (Figure

11A). However, in this case, NSCLC cells did not show higher C3 deposition when

compared to normal cells (using BEAS-2B cells as reference), except for H157 cells

(p=0.050). In addition, it was surprising that two cell lines, H460 and HTB58, showed a

significant decreased in C3 deposition compared to normal cells (pH460=0.005;

pHTB58=0.004). In view of these results, C3b inactivation was analyzed by iC3b

detection (Figure 11B). Analogous to the alternative pathway, cells showed a similar

pattern in iC3b deposition to that observed in C3 deposition. Finally, the study of C5b

deposition showed no significant differences among the two groups (Figure 11C); only

the H727 cell line showed a higher C5b deposition compared to BEAS-2B cells

(p=0.001).

Page 101: Facultad de Ciencias - CORE

Results

72

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC 3KT

HBEC 3KTp

53

HBEC

10K

T

BEA

S

0

50

100

150

200

C3 d

epositio

n (M

FI ra

tio)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC 3KT

HBEC 3KTp

53

HBEC

10K

T

BEA

S

0

50

100

150

200

iC3b d

epositio

n (M

FI ra

tio)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

BEA

S

0

1

2

3

4

5

C5 d

epositio

n (M

FI ra

tio)

∗∗ ∗∗

∗∗

∗∗

A B

C

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC 3KT

HBEC 3KTp

53

HBEC

10K

T

BEA

S

0

50

100

150

200

C3 d

epositio

n (M

FI ra

tio)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC 3KT

HBEC 3KTp

53

HBEC

10K

T

BEA

S

0

50

100

150

200

iC3b d

epositio

n (M

FI ra

tio)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

BEA

S

0

1

2

3

4

5

C5 d

epositio

n (M

FI ra

tio)

∗∗ ∗∗

∗∗

∗∗

A B

C

Figure 11. Deposition of proteins of the complement system on NSCLC (shaded bars) and normal bronchial epithelial cells (white bars) after activation of the classical complement pathway. Graphs show flow cytometry data of cells incubated in VB2+ with 10% NHS or 10% HI-NHS, as control, and stained for C3b (A), iC3b (B) and C5b (C). At least three independent experiments for each cell line were performed. Results are expressed as the ratio of the mean fluorescence intensity (MFI) between NHS and HI-NHS, and shown as mean ± SEM. Statistical differences were analyzed using BEAS cells as reference. *p<0.05; **p<0.01.

We next compared C3 deposition by the classical and the alternative pathways in

each cell line (Figure 12). The ratio of C3 deposition between both pathways was

significantly lower on NSCLC cell lines than on normal bronchial epithelial cells, using

BEAS-2B cells as reference (pA549<0.001; pH23=0.005; pH157=0.005; pH727=0.003;

pH460=0.003; pH1299=0.004 pHTB58=0.002). The reduced increase in C3 deposition on

NSCLC cell lines strongly suggests that these cells have powerful regulatory

mechanisms to inhibit complement activation on their surface.

Page 102: Facultad de Ciencias - CORE

Results

73

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

5

10

15

20

CP v

s. AP (M

FI ra

tio)

∗∗∗ ∗∗ ∗∗∗∗ ∗∗ ∗∗

∗∗

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

5

10

15

20

CP v

s. AP (M

FI ra

tio)

∗∗∗ ∗∗ ∗∗∗∗ ∗∗ ∗∗

∗∗

Figure 12. Relationship between C3 deposition after activation of the classical and alternative complement pathways (CP and AP, respectively). Alternative and classical pathway activation was performed simultaneously in each cell line and C3 deposition was analyzed. The ratios between C3 deposition in the classical and the alternative pathways were calculated. At least three independent experiments for each cell line were performed, and results are shown as mean ± SEM. Statistical differences were analyzed using BEAS cells as reference. **p<0.01; ***p<0.001.

1.3. Cell cytotoxicity due to complement activation by the alternative and

classical pathways

Complement activation leads to the assembly of the membrane attack complex

(MAC), a pore-forming agent that can penetrate into the membrane and eventually

cause cell lysis. We questioned whether NSCLC cell lines were more or less sensible

than normal bronchial epithelial cells to the lytic effect of the complement system. To

study the cytotoxicity mediated by complement activation we measured the release of

LDH to the medium after triggering complement by both pathways. A meaningless

LDH release was observed in all cells (both malignant and non-malignant) after

activation of the alternative pathway (Figure 13A). This indicates that NSCLC and

normal epithelial cell lines are resistant to the lysis induced by the spontaneous

activation of the complement system, even though NSCLC cell lines present significant

levels of C3b and C5b deposition. Regarding the classical pathway, LDH release

increased in all cell lines (Figure 13B). The cytotoxic effect due to the classical

pathway in NSCLC cell lines was comparable with that observed in BEAS-2B cells;

however, normal HBEC-KT cells showed higher sensitivity to lysis (pHBEC3KT=0.029;

pHBEC3KTp53=0.026; pHBEC10KT=0.017). This result confirms that, although lung cancer

cells activate complement, they are well-protected against its damaging effects.

Page 103: Facultad de Ciencias - CORE

Results

74

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC 3KT

HBEC 3KTp

53

HBEC 10K

T

BEAS

0

10

20

30

40

50

LDH rele

ase (%

)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC 3KT

HBEC 3KTp

53

HBEC 10K

T

BEAS

0

10

20

30

40

50

LDH rele

ase (%

)

A B∗

∗∗

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC 3KT

HBEC 3KTp

53

HBEC 10K

T

BEAS

0

10

20

30

40

50

LDH rele

ase (%

)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC 3KT

HBEC 3KTp

53

HBEC 10K

T

BEAS

0

10

20

30

40

50

LDH rele

ase (%

)

A B∗

∗∗

Figure 13. Complement-mediated cytotoxicity after stimulation of the alternative and classical pathways in NSCLC and normal epithelial cell lines. Cells were incubated with 10% NHS or HI-NHS, and cytotoxicity after activation of the alternative (A) or the classical (B) pathway was measured using an LDH release assay. The classical pathway was induced in all cell lines with an A549 antiserum. The percentage of complement-mediated lysis was calculated subtracting the percentage of lysis obtained after incubation with HI-NHS. Graphs show a representative experiment for the alternative pathway (A), and mean ± SEM of three independent experiments for the classical pathway (B). Statistical differences were analyzed using BEAS cells as reference. *p<0.05.

1.4. Expression of complement inhibitors

The fact that NSCLC cell lines present similar cytotoxicity to normal bronchial

epithelial cells, although they activate more efficiently complement, suggests

differences in the expression of complement regulators. Therefore, the expression of the

main soluble and membrane-bound complement regulators (factor H, FHL-1, factor I,

CD46, CD55 and CD59) was analyzed at the mRNA level by real time PCR (Figure

14). The expression of the inhibitors was heterogeneous throughout the cell lines, with

no apparent patterns of differential expression among groups. Another fact worthy of

mention is that the soluble regulators factor H and FHL-1 showed high levels of

expression in some NSCLC cell lines (e.i. A549, H23, H460, and HTB58). This is in

agreement with previous results from our group demonstrating that some NSCLC

tumors express and release factor H and FHL-1122, affecting tumor growth in vivo123.

We assessed the binding of factor H to the cell surface of NSCLC and normal bronchial

epithelial cells, using purified factor H or 25% HI-NHS (as a source of factor H). No

differences were seen among the two groups of cells (data not shown).

Page 104: Facultad de Ciencias - CORE

Results

75

A B

C D

E F

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0.00

0.05

0.10

0.15

FH

(G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0.0

0.1

0.2

0.3

FH

and F

HL-1

(G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0.00

0.01

0.02

0.03

0.04

FI (G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0.0

0.5

1.0

1.5

2.0

2.5

CD

46 (G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

2

4

6

8

CD

55 (G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

5

10

15

CD

59 (G

APD

H %

)

A B

C D

E F

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0.00

0.05

0.10

0.15

FH

(G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0.0

0.1

0.2

0.3

FH

and F

HL-1

(G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0.00

0.01

0.02

0.03

0.04

FI (G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0.0

0.5

1.0

1.5

2.0

2.5

CD

46 (G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

2

4

6

8

CD

55 (G

APD

H %

)

A54

9H23

H15

7

H72

7

H46

0

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10K

T

BEA

S

0

5

10

15

CD

59 (G

APD

H %

)

Figure 14. Expression of complement inhibitors by NSCLC and normal epithelial cells. mRNA semi-quantification of complement inhibitors factor H (A), FHL-1 (B), factor I (C), CD46 (D), CD55 (E) or CD59 (F) is presented as percentage relative to the GAPDH mRNA. Representative graphs for each inhibitor analysis are shown.

We next compared the concentration of factor H in bronchoalveolar lavage

samples from patients with non-malignant lung disease and from patients with lung

malignancies (both SCLC and NSCLC) (Figure 15A). Cancer patients showed

significantly higher levels of factor H in bronchoalveolar lavage samples than non-

cancer patients (p=0.002). A cytologic confirmation of the presence of malignant cells

in the clinical samples was also obtained. A ROC curve was generated for factor H. The

area under the curve was 0.79 (95% confidence interval, 0.63-0.94). A cut-off point of

Page 105: Facultad de Ciencias - CORE

Results

76

1 µg/ml was established in order to provide a sensitivity of at least 80%. Using this cut-

off, the sensitivity and specificity of the factor H test was 82% and 77%, respectively

(Figure 15B). These results were validated in an independent set of bronchoalveolar

lavage samples and in a series of sputum supernatants, with nearly identical results (data

not shown). Therefore, it can be concluded that factor H is elevated in biological fluids

from lung cancer patients and its quantification may be used as an adjunct to cytology in

the diagnosis of malignant pulmonary diseases146.

A

∗∗∗∗∗∗∗∗

Non

-mal

igna

nt

lung

dis

ease

0.0

2.5

5.0

7.5

Fa

cto

r H

(µµ µµ

g/m

l)

Lung

can

cer

Se

nsit

ivit

y

1-Specificity

BA

∗∗∗∗∗∗∗∗

Non

-mal

igna

nt

lung

dis

ease

0.0

2.5

5.0

7.5

Fa

cto

r H

(µµ µµ

g/m

l)

Lung

can

cer

Se

nsit

ivit

y

1-Specificity

B

Se

nsit

ivit

y

1-Specificity

B

Figure 15. Quantification of complement factor H in bronchoalveolar lavage supernatants A) Box plot for the quantification of factor H in bronchoalveolar lavage fluids from patients with a benign respiratory disease and patients with lung cancer. The levels of factor H were significantly higher in the lung cancer group than in the control group (**p<0.01). B) A ROC curve was generated for factor H. Area under the curve was 0.79 (95% confidence interval, 0.63-0.94; p=0.003).

Following with the characterization of the membrane-bound regulators in

NSCLC, the role of regulators of the MAC formation was more deeply studied. We first

analyzed the expression of the main inhibitor at this level, CD59. Our intention was to

study the inhibitory effect of CD59 on the cell membrane before and after the treatment

with phosphatidylinositol-specific phospholipase C (PI-PLC). CD59, as well as CD55,

is anchored in the plasma membrane via a conserved glycan-phosphatidylinositol (GPI)

anchor. The anchor consists of a phosphatidylinositol group and a carbohydrate-

containing linker that binds to the C-terminal amino acid of a mature protein. This type

of binding is shared by a heterogeneous group of proteins and can be released from cell

membranes by purified bacterial PI-PLCs. The first observation we made was that

CD59 protein expression on the cell membrane was irregular among NSCLC and

normal epithelial cells (Figure 16A), as expected from our previous results in mRNA

Page 106: Facultad de Ciencias - CORE

Results

77

expression. We next analyzed the remaining CD59 on the cell membrane after PI-PLC

treatment. The reason to perform this analysis was that, in some cases, palmitate, one of

the two fatty acids within the hydrophobic phosphatidylinositol group, is esterified to

the 2-OH group of myo-inositol; this feature prevents the release of GPI-proteins after

PI-PLC action and distinguishes two types of proteins, PI-PLC resistant and PI-PLC

sensible157. We noticed that both the sensitivity and resistance to PI-PLC treatment were

commonly present in the two cell groups (Figure 16B). When the remaining CD59 in

the cells was compared to that on BEAS-2B cells, four NSCLC cell lines showed to be

more resistant to PI-PLC action (pA549=0.028; pH23=0.021; pH460=0.014; pH1299=0.023),

although HBEC10KT cells also showed this resistance to the treatment

(pHBEC10KT=0.019). After activation of the alternative complement pathway, C5b-9

deposition on the cell membrane increased in cells treated with PI-PLC compared to

untreated cells; however, no correlation with the levels of CD59 that remained attached

to the cell was observed (data not shown). These results prompted us to look for

alternative regulatory mechanisms. Casein kinase 2 (CK2), an ubiquitous and

constitutively protein kinase that is expressed on the cell membrane, regulates MAC

assembly. We analyzed the expression of CK2 on the cell membrane of tumor and

normal epithelial cells. However, no difference in CK2 expression on the cell surface

was uncovered (Figure 16C).

Page 107: Facultad de Ciencias - CORE

Results

78

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10 KT

BEA

S

0

500

1000

1500

2000

2500

CD

59 e

xpre

ssio

n (M

FI units)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10 KT

BEA

S

0

20

40

60

80

100

CD

59 a

fter

PI-PLC

(%

)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10 KT

BEA

S

0

5

10

15

20

CK

2 e

xpre

ssio

n (M

FI units)

A B

C

∗∗

∗ ∗∗

∗∗

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10 KT

BEA

S

0

500

1000

1500

2000

2500

CD

59 e

xpre

ssio

n (M

FI units)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10 KT

BEA

S

0

20

40

60

80

100

CD

59 a

fter

PI-PLC

(%

)

A54

9H23

H15

7

H46

0

H72

7

H12

99

HTB

58

HBEC

3KT

HBEC

3KTp

53

HBEC

10 KT

BEA

S

0

5

10

15

20

CK

2 e

xpre

ssio

n (M

FI units)

A B

C

∗∗

∗ ∗∗

∗∗

Figure 16. Expression of CD59 and ecto-phosphokinase CK2 on NSCLC and normal epithelial cells. CD59 expression (A), remaining CD59 on the cell membrane after the treatment with PI-PLC (B), and CK2 expression (C) were measured by flow cytometry. Data represent mean fluorescence intensity (MFI) from three independent experiments and are expressed as mean ± SEM. *p<0.05; **p<0.01.

1.5. Effect of hypoxia in the expression of complement inhibitors on NSCLC

cells

In a previous work, Drs Anna Blom and Marcin Okroj from the University of

Lund (Malmö, Sweeden), in collaboration with our group, had shown that two NSCLC

cell lines, H2087 and H358, express not only the whole set of soluble complement

inhibitors (factor H, C4BP and factor I), but also the main membrane-bound

complement inhibitors (CD46, CD55 and CD59)124. This work prompted us to study the

effect of different physiological conditions in complement regulation. In particular, we

examined how hypoxic conditions influence the expression of complement

regulators158. For the study, H358 and H2087 cells were incubated in hypoxia (48

hours), in hypoxia followed by normoxia (24 hours each) or in normoxia (48 hours).

Expression of complement inhibitors, at mRNA and protein levels, was analyzed. Real-

time PCR showed a significant reduction in the expression of soluble and membrane

inhibitors in both cell lines, except for CD59 (Figure 17A). CD59 levels on the cell

membrane, analyzed by flow cytometry, were not affected by hypoxic conditions. CD55

Page 108: Facultad de Ciencias - CORE

Results

79

exhibited a significant decrease in its expression after 24 h of hypoxia followed by

reoxygenation in H358 cells. CD46 presented a substantial loss in both cell lines after

24 h of hypoxia/reoxygenation, which was intensified after 48 h of hypoxia (Figure

17B). Factor I and factor H levels were measured by ELISA. Conditioned media

collected from both cell lines showed a strong decrease in the levels of the soluble

factors in hypoxia/reoxygenation and hypoxia, in comparison to normoxic conditions

(Figure 17C). These results imply that under hypoxic conditions NSCLC cells give up

some of their defense mechanisms and become more prone to complement attack.

CFH CFI CD46 CD55 CD590

50

100

150

mR

NA e

xpessio

nvs. norm

oxic

cells

(%

)

CFH CFI CD46 CD55 CD590

50

100

150

mR

NA e

xpessio

nvs. norm

oxic

cells

(%

)

CD46 CD55 CD590

50

100

150

Pro

tein

expre

ssio

nvs. norm

oxic

cells

(%

)

CD46 CD55 CD590

50

100

150

Pro

tein

expre

ssio

nvs. norm

oxic

cells

(%

)

FH FI0

50

100

150

Pro

tein

rele

ased

vs. norm

oxic

cells

(%

)

FH FI0

50

100

150

Pro

tein

rele

ased

vs. norm

oxic

cells

(%

)

H358 cells H2087 cells

A

B

C

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗

∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗ ∗∗∗ ∗∗∗ ∗∗∗ ∗∗∗

Normoxia 24h Hypoxia/Reoxygenation 48h Hypoxia

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

CFH CFI CD46 CD55 CD590

50

100

150

mR

NA e

xpessio

nvs. norm

oxic

cells

(%

)

CFH CFI CD46 CD55 CD590

50

100

150

mR

NA e

xpessio

nvs. norm

oxic

cells

(%

)

CD46 CD55 CD590

50

100

150

Pro

tein

expre

ssio

nvs. norm

oxic

cells

(%

)

CD46 CD55 CD590

50

100

150

Pro

tein

expre

ssio

nvs. norm

oxic

cells

(%

)

FH FI0

50

100

150

Pro

tein

rele

ased

vs. norm

oxic

cells

(%

)

FH FI0

50

100

150

Pro

tein

rele

ased

vs. norm

oxic

cells

(%

)

H358 cells H2087 cells

A

B

C

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗

∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗ ∗∗∗ ∗∗∗ ∗∗∗ ∗∗∗

Normoxia 24h Hypoxia/Reoxygenation 48h HypoxiaNormoxia 24h Hypoxia/Reoxygenation 48h Hypoxia

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

∗∗∗∗∗∗∗∗∗∗∗∗

Figure 17. Expression of complement inhibitors by NSCLC cells in hypoxic conditions. Expression in normoxic cells was set as 100% (indicated by a dotted line). Data from at least three experiments are summarized in the graphs and expressed as mean ± SEM. A) mRNA expression of each complement inhibitor was studied by real-time PCR and calculated as percentage relative to the GAPDH mRNA. B) Presence of membrane-bound complement inhibitors, CD46, CD55 and CD59, on NSCLC cells exposed to hypoxia were analyzed by flow cytometry, and expressed as percent of mean fluorescence intensity (MFI) versus normoxic cells. C) Concentrations of soluble factors were measured by ELISA. *p<0.05; **p<0.01; ***p<0.001.

Page 109: Facultad de Ciencias - CORE

Results

80

1.6. C5a levels in patients with NSCLC

Our in vitro data suggest that NSCLC cells are able to activate complement

spontaneously, leading to the deposition of complement proteins on the cell membrane

and, likely, to the release of complement components, such as anaphylatoxins, to the

extracellular medium. To assess whether this may be the case in vivo, we evaluated the

concentration of C5a, the most potent anaphylatoxin, in clinical samples from patients

diagnosed with lung cancer. We compared it with the concentration of this protein in

samples from control patients. Quantification of C5a was first performed in plasma

from NSCLC (both adenocarcinoma and squamous cell carcinoma) patients and healthy

donors matched by age and smoking history (Figure 18A). Cancer patients showed

significantly higher levels of C5a in plasma than healthy donors (9.62 ± 2.15 ng/ml vs.

5.13 ± 1.15 ng/ml; p<0.001). No histological differences were found. C5a was also

quantified in bronchoalveolar lavage supernatants from patients with non-malignant

lung disease and patients with lung malignancies (both SCLC and NSCLC) (Figure

18B). C5a levels in cancer patients were significantly higher than in patients with non-

malignant respiratory diseases (40.16 ± 37.08 ng/ml vs. 25.69 ± 25.79 ng/ml; p=0.049).

Again, no statistical differences were observed between histological subtypes.

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

Con

trol

NSC

LC

0

10

20

30

40

C5a (ng/m

l)

Con

trol

Lung

can

cer

0

50

100

150

200

C5a (ng/m

l)

A B

∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗∗

Con

trol

NSC

LC

0

10

20

30

40

C5a (ng/m

l)

Con

trol

Lung

can

cer

0

50

100

150

200

C5a (ng/m

l)

A B

Figure 18. C5a concentration in biological fluids from patients with lung cancer and controls. A) Box plot for the quantification of C5a in plasma samples from patients with NSCLC or from healthy donors. B) Box plot for the quantification of C5a in bronchoalveolar lavage supernatants from patients with lung cancer (NSCLC and SCLC) and patient with non-malignant respiratory diseases. *p<0.05; ***p<0.001.

Page 110: Facultad de Ciencias - CORE

Results

81

2. COMPLEMENT DEFICIENCY IMPAIRS TUMOR GROWTH

Our in vitro experiments have shown that lung tumor cells activate complement

spontaneously; however this activation is not able to damage them. Most probably, this

is due to the acquisition of complement regulatory mechanisms which inhibit the

progression of complement activation before it causes harm to the cells. At this point,

we speculated with the possibility that this partial complement activation may have

other functional consequences to the cell or its microenvironment. In particular, the

anaphylatoxin C5a, overexpressed in biological fluids from lung cancer patients, have

been shown to have promoting effects on tumor cells, and on cells present within the

tumor microenvironment. Bearing this fact in mind, the next step was to evaluate the in

vivo consequences of complement deprivation, to check if complement activation could

be beneficial for lung tumorigenesis. We used C3 deficient mice and the

pharmacological blockade of C5aR-1 (the main C5a receptor).

A syngeneic model of lung cancer was used to perform the in vivo experiments.

The model consisted in the subcutaneous injection of Lewis lung carcinoma (LLC or

3LL) cells into the flanks of C57BL/6J mice. We first confirmed that 3LL cells were

able to activate the alternative pathway of complement in vitro. Cells incubated with

10% normal mouse serum (NMS) presented higher C3 deposition than cells incubated

with 10% heat-inactivated NMS (Figure 19A).

2.1. Inhibition of tumor growth by C3 deficiency

The formation of the C3 convertase is a main step in complement activation. At

this point, the three central pathways converge. Consequently, C3 deficiency is a good

manner to prevent the generation of complement effectors. Therefore, using C3

knockout mice we were able to study the effect of complement, in a wide and general

way, in the promotion of tumors.

Before starting the experiments, mice were genotyped and C3 expression was

checked. The presence of C3 gene was evaluated by PCR and the concentration of

serum C3 by ELISA. The assays confirmed that C3-deficient mice lacked the C3 gene

and had undetectable concentrations of C3 in their serum (data not shown).

We next assessed tumor growth in C3-deficient and wild-type mice after

subcutaneous inoculation of 3LL cells. Tumor growth was significantly impaired in the

Page 111: Facultad de Ciencias - CORE

Results

82

absence of C3 (Figure 19B). Tumor volumes measured every three days, starting about

one week after subcutaneous inoculation of tumor cells, were significantly lower in the

C3-deficient mice than in the wild-type controls from the 15th day after cell injection

(p=0.002) until the end of the experiment (p=0.009). At the end of the experiment, we

observed no increase in C3 levels in wild-type tumor-bearing mice when compared with

a pool of serum from ten tumor-free wild-type mice (data not shown). This implies that

these tumor cells are not producing detectible C3 to reverse its deficiency in C3

knockout mice. This result indicates that complement activation promotes lung tumor

growth.

It has been reported that C5 can be proteolyzed into C5a and C5b in the genetic

absence of C334. If that was the case in our in vivo model, this would imply that C5a and

C5b may be present in C3 deficient mice and would not be responsible for the tumor

promoting effect of complement. Moreover, an ELISA for murine C5a showed similar

levels of the anaphylatoxin in C3-deficient and wild-type animals (data not shown). To

rule out any possible effect of C3-independent C5 convertases in our model, we

repeated the last experiment using C3 deficient animals injected with cobra venom

factor (CVF). Injection of CVF has been widely used to deplete complement in several

animal models. CVF resembles functionally and structurally C3b; this characteristic

allows it to bind factor B and to form a stable C3/C5 convertase that cleaves both

complement components C3 and C5. The uncontrollable activity of the convertase

induces consumption of complement proteins C3 and C5, resulting in complement

depletion159. Therefore, the injection of CVF in C3 deficient animals guaranteed a

complete depletion of C5 from circulation.

To settle the timing of CVF injection, we monitored C3 levels in wild-type mice

after a dose of 5 µg CVF. C3 levels were undetectable during three days after the

injection and started to recover after the forth day. After one week of injection, C3

levels became normal again (Figure 19C). Based on these results, we decided to inject

CVF in C3 deficient animals at a dose of 5 µg every three days. Using this regiment of

administration, we again assessed in vivo tumor growth in C3-deficient mice after

subcutaneous inoculation of 3LL tumor cells. As previously described, cell growth was

significantly reduced in C3-deficient animals. This difference was maintained in C3-

deficient mice injected with CVF (Figure 19D).

Page 112: Facultad de Ciencias - CORE

Results

83

0

1000

2000

3000

4000

9 12 15 18 21 24

WT

C3 KO

0

1000

2000

3000

4000

9 12 15 18 21 24

WT

C3 KO

C3 KO + CVF

∗∗

∗∗

A B

C D

Tu

mo

r vo

lum

e (

mm

3)

Tu

mo

r vo

lum

e (

mm

3)

100 101 102 103 104

Fluorescence intensity

025

0C

ell

nu

mb

er

Basal 1 2 3 4 5 6 7 8

50

100

150

Basal 1 2 3 4 5 6 7 8 Days after CVF injection

Basal 1 2 3 4 5 6 7 8

50

100

150

Basal 1 2 3 4 5 6 7 8 Days after injection

C3 v

sb

asal

valu

e (

%)

∗∗

n.s.

Days after injection

0

1000

2000

3000

4000

9 12 15 18 21 24

WT

C3 KO

0

1000

2000

3000

4000

9 12 15 18 21 24

WT

C3 KO

C3 KO + CVF

∗∗

∗∗

A B

C D

Tu

mo

r vo

lum

e (

mm

3)

Tu

mo

r vo

lum

e (

mm

3)

100 101 102 103 104

Fluorescence intensity

025

0C

ell

nu

mb

er

100 101 102 103 104

Fluorescence intensity

025

0C

ell

nu

mb

er

Basal 1 2 3 4 5 6 7 8

50

100

150

Basal 1 2 3 4 5 6 7 8 Days after CVF injection

Basal 1 2 3 4 5 6 7 8

50

100

150

Basal 1 2 3 4 5 6 7 8 Days after injection

C3 v

sb

asal

valu

e (

%)

∗∗

n.s.

Days after injection

Figure 19. Tumor growth of Lewis lung carcinoma (3LL) cells injected in C57BL mice with normal or deficient complement activity. A) Activation of the alternative pathway of complement in 3LL cells measured by flow cytometry. Cells were incubated with normal mouse serum (white peak) or heat-inactivated normal mouse serum (shaded peak). B) Tumor growth of the 3LL cell line injected s.c. in C3-deficient mice (C3 KO) and wild-type mice (WT). Significant differences were observed from day 15th after injection. Data are representative of three independent experiments with at least 10 mice per group. Tumor volume is expressed as mean ± SEM. C) Consumption of serum C3 in C57BL mice after injection of three i.p. doses of 5 µg CVF; doses were injected 28, 24 and 4 hours before the collection of the first blood sample till the 8th day. C3 concentration in serum was determined by ELISA and expressed as percentage of basal C3 in mice without CVF treatment. D) Tumor growth of the 3LL cell line injected in wild-type and C3-deficient mice treated with or without CVF. Data correspond to one experiment with at least 10 mice per group. Tumor volume is expressed as mean ± SEM. *p<0.05; **p<0.01.

2.2. Inhibition of tumor growth by C5aR-1 blockade

We postulated that the C5a anaphylatoxin may be a good candidate for the tumor

promoting effect of complement activation. C5a causes a number of biological effects,

which are essential in the primary innate immune response160. However, it has been

demonstrated that the functions of C5a are not limited to the immune response. C5a also

elicits several cellular responses related to tumor growth and progression. In addition,

we observed increased levels of C5a in plasma and bronchioalveolar lavage fluid from

patients with NSCLC. These data, together with our results in C3-deficient mice,

directed us towards the study of the implications of C5a on tumor growth in our

syngeneic model of lung cancer. To achieve this purpose, C5aR-1 was blocked with the

Page 113: Facultad de Ciencias - CORE

Results

84

cyclic hexapeptide AcF(OP(D)ChaWR). Tumor growth was compared between wild-

type animals treated every three days with the C5aR antagonist and wild-type animals

treated with the control cyclic hexapeptide AcF(OP(D)ChaA(D)R). The blockade of

C5aR-1 decreased tumor growth, relative to the effect of the control peptide (Figure

20A). This reduction reached statistical significance from the 24th day of tumor cell

injection (p=0.035) to the end of the experiment (p=0.005). The pharmacological

blockade of C5aR-1 was also used in animals with C3-deficiency. We questioned

whether blocking the C5a receptor in animals deficient in C3 had an additive effect in

tumor growth. For this purpose, C3-deficient mice were treated with the C5aR-1

antagonist or with the control peptide after injection of 3LL cells. Tumor growth was

compared with wild-type mice treated with the control peptide. Tumor growth was

reduced in C3 knockout mice independently of the C5aR-1 blockade (Figure 20B). As

with the CVF studies, these results suggest that no C3-independent C5 convertases are

generated in the C3 knockout model which may have affected the interpretation of the

results. More importantly, our results demonstrate the participation of C5aR-1 in lung

tumor growth in vivo.

0

500

1000

1500

2000

2500

12 15 18 21 24 27 30 32

WT + control peptide

WT + C5aR antagonist

0

500

1000

1500

2000

2500

12 15 18 21 24 27 30 32

WT + control peptide

C3 KO + control peptide

C3 KO + C5aR-1 antagonist∗∗

n.s.

Tu

mo

r vo

lum

e (

mm

3)

Days after injection

Tu

mo

r vo

lum

e (

mm

3)

Days after injection

∗∗

A B

0

500

1000

1500

2000

2500

12 15 18 21 24 27 30 32

WT + control peptide

WT + C5aR antagonist

0

500

1000

1500

2000

2500

12 15 18 21 24 27 30 32

WT + control peptide

C3 KO + control peptide

C3 KO + C5aR-1 antagonist∗∗

n.s.

Tu

mo

r vo

lum

e (

mm

3)

Days after injection

Tu

mo

r vo

lum

e (

mm

3)

Days after injection

∗∗

A B

Figure 20. Pharmacological blockade of C5aR-1 decreases tumor growth. A) Tumor volumes of 3LL cells in wild-type mice treated with the C5aR-1 antagonist or the control peptide. B) Tumor volumes in wild-type or C3-deficient animals treated with the C5aR-1 antagonist or the control peptide. In each case, data are representative of two independent experiments, each one with at least 10 mice per group. Tumor volume is expressed as mean ± SEM. *p<0.05; **p<0.01.

Page 114: Facultad de Ciencias - CORE

Results

85

3. COMPLEMENT ACTIVATION REGULATES ANTITUMOR

IMMUNE RESPONSE

Once we had demonstrated that the activation of C5aR-1 is important for tumor

growth in vivo, we were interested in evaluating the mechanisms by which this effect

takes place. Immune system activation is a crucial mechanism that contributes to the

regulation of tumor growth, and C5a is a modulator of the immune response. Therefore,

it was likely that C5a was mediating some sort of control in the immune response that

would explain its role in tumor growth. Using our syngeneic animal model of lung

cancer, we analyzed the effect of C3 deficiency and C5aR-1 blockade in the immune

response to the tumor.

3.1. Immune activation is induced in C3-deficient animals

Immune cells within the spleens of animals from the experiment described in

section 2.1. (in which C3 deficient and wild-type mice were inoculated subcutaneously

with 3LL cells) were analyzed by flow cytometry. We observed that complement

deficiency quantitatively altered the populations of cells present in the spleen. As shown

in Figure 21, animals deficient in C3 (which presented smaller tumors than wild-type

ones) exhibited important changes in some immune populations. There was an increase

of CD19+ (p=0.004) and CD8+ (p=0.017) populations. There was also a tendency to the

increase of CD4+ cells, although the difference did not reach statistical significance

(p=0.099). In addition, myeloid-derived suppressor cells (MDSCs; CD11b+Ly6c+)

decreased in C3 knockout mice (p=0.022). MDSCs consist of two major

subpopulations, granulocytic and monocytic, both differ in the expression of two

epitopes, Ly6C and Ly6G, in the GR1 protein. The Ly6G molecule is expressed

primarily on granulocytes161, whereas Ly6C is typically expressed on monocytes162.

Using an antibody against Ly6C we differentiated granulocytic MDSCs, with Ly6Clow,

from monocytic MDSCs, with Ly6Chigh. We observed that both subpopulations were

reduced in the spleens of C3 knockout mice compared to those in the spleens of wild-

type ones (pLy6C-low= 0.036; pLy6C-high= 0.007). We also analyzed the population of Treg

cells (CD4+CD25+Foxp3+) within the spleen. The percentage of Treg cells tended to

Page 115: Facultad de Ciencias - CORE

Results

86

decrease in C3 knockout mice, but the changes were not statistically significant.

However, the ratio between Treg and CD4 cells (Treg %/CD4 %) was clearly different

between groups (p<0.001). These results indicate that complement sufficient animals

present an immune suppressive microenvironment that may tolerate tumor progression.

WT

C3 KO

0

10

20

30

40

50

CD

19 (

%)

WT

C3 KO

0

5

10

15

CD

4 (%

)

WT

C3 KO

0

5

10

15

CD

8 (%

)

WT

C3 KO

0

1

2

3

Tre

gs (

%)

WT

C3 KO

0

5

10

15

20

Tre

gs (

%)

/ C

D4 (%

)

WT

C3 KO

0

2

4

6

8

10

MD

SC

(Ly6c low

) (%

)

WT

C3 KO

0.0

0.5

1.0

1.5

2.0

MD

SC

(Ly6c h

igh) (%

)

WT

C3 KO

0

5

10

15

Tota

l M

DS

C (%

)

∗∗ns

∗∗ ∗∗

∗∗∗ns

WT

C3 KO

0

10

20

30

40

50

CD

19 (

%)

WT

C3 KO

0

5

10

15

CD

4 (%

)

WT

C3 KO

0

5

10

15

CD

8 (%

)

WT

C3 KO

0

1

2

3

Tre

gs (

%)

WT

C3 KO

0

5

10

15

20

Tre

gs (

%)

/ C

D4 (%

)

WT

C3 KO

0

2

4

6

8

10

MD

SC

(Ly6c low

) (%

)

WT

C3 KO

0.0

0.5

1.0

1.5

2.0

MD

SC

(Ly6c h

igh) (%

)

WT

C3 KO

0

5

10

15

Tota

l M

DS

C (%

)

∗∗∗∗nsns

∗∗

∗∗∗∗ ∗∗∗∗

∗∗∗∗∗∗nsns

Figure 21. C3 deficiency affects populations of splenocytes in tumor-bearing mice. Graphs represent data from the flow cytometry analysis of splenocytes obtained from wild-type (WT) or C3 knockout mice (C3 KO). Splenocytes were stained with specific antibodies to CD19+, CD4+, CD8+, CD11b+Ly6c+ (MDSC), and CD4+CD25+Foxp3+ (Tregs). Individual results per animal and mean per group are shown. *p<0.05; **p<0.01; ***p<0.001.

Page 116: Facultad de Ciencias - CORE

Results

87

3.2. C5aR-1 blockade induces immune activation

Due to the last result, and considering that C5a has been proved to be a key factor

in the immune regulation of tumors, mainly by the recruitment of MDSCs107; we

analyzed the splenocytes from tumor-bearing animals treated with the C5aR-1

antagonist or the control peptide. Similarly to the results obtained in C3 knockout mice,

splenocytes from animals treated with the control peptide showed an

immunosuppressive microenvironment compared to the animals treated with the C5aR-

1 antagonist (Figure 22). In this case, the increased in CD19+, CD4+ and CD8+

populations with C5aR-1 blockade did not reach significance, although a tendency was

observed. Total MDSCs, as well as MDSC subpopulations, were significantly reduced

when C5aR-1 was blocked (pTotal= 0.015; pLy6C-low=0.018; pLy6C-high= 0.045). Finally,

there were no differences among groups in the percentage of Treg cells or the ratio Treg

%/CD4 %.

Page 117: Facultad de Ciencias - CORE

Results

88

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

10

20

30

CD

19 (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

2

4

6

8

10

CD

4 (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

5

10

15

CD

8 (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0.0

0.5

1.0

1.5

2.0

Tre

gs (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

5

10

15

20

Tre

gs (%

) / C

D4 (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

5

10

15

20M

DS

C (Ly6c low

) %

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

1

2

3

4

MD

SC

(Ly6c h

igh) %

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

5

10

15

20

MD

SC

%

ns

nsns

∗ ∗

nsns

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

10

20

30

CD

19 (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

2

4

6

8

10

CD

4 (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

5

10

15

CD

8 (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0.0

0.5

1.0

1.5

2.0

Tre

gs (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

5

10

15

20

Tre

gs (%

) / C

D4 (%

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

5

10

15

20M

DS

C (Ly6c low

) %

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

1

2

3

4

MD

SC

(Ly6c h

igh) %

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

0

5

10

15

20

MD

SC

%

nsns

∗∗

nsnsnsns

∗∗ ∗∗

nsnsnsns

Figure 22. C5aR-1 blockade affects populations of splenocytes in tumor-bearing mice. Graphs represent data from the flow cytometry analysis of splenocytes obtained from mice treated with the control peptide or the C5aR-1 antagonist. Splenocytes were stained with specific antibodies to CD19+, CD4+, CD8+, CD11b+Ly6c+ (MDSC), and CD4+CD25+Foxp3+ (Tregs). Individual results per animal and mean per group are shown. *p<0.05.

Page 118: Facultad de Ciencias - CORE

Results

89

3.3. C3 deficiency and C5aR-1 blockade reduces the expression of

immunosuppressive cytokines within the tumor

Analysis of splenocytes is a valuable information about the immune response

generated against the tumor, but direct evaluation of the tumor microenvironment is also

needed. For this purpose, total RNA was extracted from explanted tumors of wild-type

animals treated with the control peptide or the C5aR-1 antagonist and from C3 deficient

mice treated with the control peptide. In collaboration with Dr. Lasarte and Dr. Riezu

(Division of Gene Therapy and Hepathology, CIMA), we performed real time PCR for

twenty four molecules related to the immune response. The study showed that

expression of PDL1, CTLA4, LAG3 and ARG1 significantly decreased in tumors from

animals treated with the C5aR-1 antagonist and in tumors from C3-deficient animals

(Figure 23). IL10, IL6 and IL12p35 expression decreased in tumors from animals

treated with the antagonist; and CD70 in tumors from C3-deficient mice. The other

molecules studied (IDO, TNF-α, IL-2, IL-4, Foxp3, PD1, CCL17, CCL22, GITR, TGF-

β, INF-γ, OX40L, MCP1, granzyme A, granzyme B and perforin) showed similar

expression levels between the different groups (data not shown).

Page 119: Facultad de Ciencias - CORE

Results

90

ns∗∗

∗∗

∗∗

∗∗∗ ∗

ns

ns ∗

ns

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.000

0.005

0.010

0.015

0.020IL

10 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.0

0.2

0.4

0.6

0.8

PD

L1 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.00

0.02

0.04

0.06

0.08

0.10

CTLA4 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 ant

agon

ist

C3 KO + con

trol pep

tide

0

2

4

6

LAG

3 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0

5

10

15

20

AR

G1 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.0000

0.0005

0.0010

0.0015

IL12p35 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.00

0.01

0.02

0.03

0.04

IL6 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 ant

agon

ist

C3 KO + con

trol pep

tide

0.0000

0.0002

0.0004

0.0006

0.0008

0.0010

CD

70 m

RN

A (

ββ ββ-a

ctin %

)

∗∗

nsns∗∗∗∗

∗∗

∗∗∗∗

∗∗

∗∗∗∗

∗∗∗∗∗∗ ∗∗

∗∗

∗∗

nsns

∗∗

nsns ∗∗

nsns

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.000

0.005

0.010

0.015

0.020IL

10 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.0

0.2

0.4

0.6

0.8

PD

L1 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.00

0.02

0.04

0.06

0.08

0.10

CTLA4 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 ant

agon

ist

C3 KO + con

trol pep

tide

0

2

4

6

LAG

3 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0

5

10

15

20

AR

G1 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.0000

0.0005

0.0010

0.0015

IL12p35 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.00

0.01

0.02

0.03

0.04

IL6 m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 ant

agon

ist

C3 KO + con

trol pep

tide

0.0000

0.0002

0.0004

0.0006

0.0008

0.0010

CD

70 m

RN

A (

ββ ββ-a

ctin %

)

Figure 23. RNA expression in 3LL tumors of molecules related to the immune response. Total mRNA was extracted from tumors of wild-type mice (WT) treated with the control peptide or the C5aR-1 antagonist, and from tumors of C3 knockout mice (C3 KO) treated with the control peptide. mRNA levels of the different immune mediators were calculated by real time PCR and expressed as percentage relative to the β-actin mRNA in each tumor. Individual results per animal and mean per group are shown. *p<0.05; **p<0.01; *** p<0.001.

Page 120: Facultad de Ciencias - CORE

Results

91

4. EFFECT OF C5a IN THE PROMOTION OF ANGIOGENESIS

At this point of the study, we can conclude that lung cancer cells activate

complement and that this activation is important for tumor growth, because released

factors such as C5a generate an immunosupressive microenvironment. Nevertheless, the

relevance of C5a may not be limited to the regulation of immune response. In this sense,

it has been suggested that C5a may affect the endothelium during inflammation.

Therefore, our next step in the study was to evaluate the effect of C5a in human

umbilical vein endothelial cell (HUVEC) proliferation and migration, as well as the

capacity of C5a to induce angiogenesis.

4.1. Effect of C5a on HUVEC proliferation

First, the expression of C5aR-1 on HUVECs was demonstrated by flow

cytometric analysis (Figure 24A). Cell proliferation assays were performed in HUVECs

stimulated with different concentrations of C5a. At first, a wide range of C5a

concentrations was used, and proliferation was measured after 96 hours. In these

conditions, C5a did not affect the proliferation of HUVECs (Figure 24B). Next, we

tested the capacity of C5a to synergize the proliferative activity of bFGF, since this

molecule is one of the supplements needed for HUVEC growth. To select the

appropriate bFGF concentration, we performed a dose-response assay where HUVECs

were treated with bFGF at different concentrations, ranging from 0.02 to 10 ng/ml

(Figure 24C). We selected 0.31 ng/ml of bFGF because this was one of the lowest

concentrations in the linear slope of the dose-response curve sufficient to support cell

proliferation. MTT assays were performed adding bFGF at 0.31 ng/ml in two different

conditions: C5a at different concentrations ranging from 12.5 to 100 ng/ml, or C5a at a

constant concentration of 100 ng/ml, with the other two anaphylatoxins, C3a and C4a,

at 25, 50 or 100 ng/ml. None of these attempts had any effect in HUVEC proliferation

(Figure 24D).

Page 121: Facultad de Ciencias - CORE

Results

92

contro

l

FGF (10 ng

/ml)

0.5 5 25 50 25

050

010

00

0.0

0.1

0.2

0.3

0.4

0.5

C5a (ng/ml)

Absorb

ance (540 n

m)

0.0

0.1

0.2

0.3

Absorb

ance (540 n

m)

00.02

0.04

0.08

0.15

0.31

0.62

1.25

2.5 5 10

0.0

0.1

0.2

0.3

0.4

bFGF (ng/ml)

Absorb

ance (540 n

m)

100 101 102 103 104

20

00

bFGF (ng/ml)

C5a (ng/ml)

C3a and C4a (ng/ml)

- 10 0.31 0.31 0.31 0.31 0.31 0.31 0.31 0.31

- - - 12.5 25 50 100 - - -

- - - - - - - 25 50 100

A B

C D

Fluorescence intensity

Cell

nu

mb

er

contro

l

FGF (10 ng

/ml)

0.5 5 25 50 25

050

010

00

0.0

0.1

0.2

0.3

0.4

0.5

C5a (ng/ml)

Absorb

ance (540 n

m)

0.0

0.1

0.2

0.3

Absorb

ance (540 n

m)

00.02

0.04

0.08

0.15

0.31

0.62

1.25

2.5 5 10

0.0

0.1

0.2

0.3

0.4

bFGF (ng/ml)

Absorb

ance (540 n

m)

100 101 102 103 104

20

00

100 101 102 103 104

20

00

bFGF (ng/ml)

C5a (ng/ml)

C3a and C4a (ng/ml)

- 10 0.31 0.31 0.31 0.31 0.31 0.31 0.31 0.31

- - - 12.5 25 50 100 - - -

- - - - - - - 25 50 100

A B

C D

Fluorescence intensity

Cell

nu

mb

er

Figure 24. Expression of C5aR-1 in human umbilical vein endothelial cells (HUVECs) and effect of C5a on HUVEC proliferation. A) Detection of C5aR-1 on the cell membrane of HUVECs was assessed by flow cytometry. Cells were incubated with an anti-C5aR-1 antibody (white peak) or with an isotype control (shaded peak). B) Proliferation assay with cells incubated with C5a at different concentrations in basal medium for 96 hours. C) Proliferation assay with bFGF at different concentrations in basal medium. D) Proliferation assay with combinations prepared with 0.31 ng/ml bFGF and C5a at different concentrations (black bars), or 0.31 ng/ml bFGF with 100 ng/ml C5a and the other two anaphylatoxins, C3a and C4a, at different concentrations (grey bars). Negative and positive controls in B and D were done with basal medium and with 10 ng/ml bFGF, respectively. In all graphs, one representative experiment is shown, and results are expressed as the mean optical density of the MTT assay ± SEM.

4.2. Effect of C5a on HUVEC migration

We next investigated the capacity of C5a to induce HUVEC migration using the

transwell chamber assay. HUVECs were plated on transwells and were stimulated with

different concentrations of C5a, ranging from 0.03 to 500 ng/ml, during 48 hours.

HUVECs showed a bell shaped dose-response (Figure 25A and 25B). A significant

increase in migration, compared to the untreated cells, was observed within 0.125 and

5 ng/ml of C5a (p0.125=0.015; p0.25<0.001; p0.5=0.012; p5<0.001).

Page 122: Facultad de Ciencias - CORE

Results

93

cont

rol

0.03

0.06

0.12

50.25 0.

5 5 50 500

0

20

40

60

80

100

C5a (ng/ml)

Cell

num

ber

/ field

∗∗∗

∗∗∗

Control

C5a 0.125 ng/ml C5a 0.25 ng/ml

C5a 0.5 ng/ml C5a 5 ng/ml

A B

cont

rol

0.03

0.06

0.12

50.25 0.

5 5 50 500

0

20

40

60

80

100

C5a (ng/ml)

Cell

num

ber

/ field

∗∗∗

∗∗∗

cont

rol

0.03

0.06

0.12

50.25 0.

5 5 50 500

0

20

40

60

80

100

C5a (ng/ml)

Cell

num

ber

/ field

∗∗∗

∗∗∗

cont

rol

0.03

0.06

0.12

50.25 0.

5 5 50 500

0

20

40

60

80

100

C5a (ng/ml)

Cell

num

ber

/ field

∗∗∗

∗∗∗

Control

C5a 0.125 ng/ml C5a 0.25 ng/ml

C5a 0.5 ng/ml C5a 5 ng/ml

A B

Figure 25. Effect of C5a on HUVEC migration in the transwell assay. C5a was added at different concentrations in basal medium on the lower side of the transwells. HUVECs were plated in the upper chamber in basal medium. At 48 hours after plating, cells that had migrated to the underside of the transwells were fixed and stained with crystal violet. A) Cell migration in each transwell was analyzed by counting cells in eight different fields of the transwell and calculating the mean value. Data are collected from three independent experiments and shown as mean ± SEM. *p<0.05; ***p<0.001. B) Representative images of HUVEC migration at different concentrations are shown at x10 magnification.

Page 123: Facultad de Ciencias - CORE

Results

94

4.3. Effect of C5a on HUVEC tube formation

In the presence of angiogenic compounds, HUVECs can form tube-like vessels on

Matrigel, a commercial basement membrane matrix derived from the Engelbreth-Holm-

Swarm mouse sacoma163. With this assay, we tested the potential of C5a to induce

angiogenesis in vitro. HUVECs were plated on gelled Matrigel with 10 ng/ml C5a in

EBM-2 medium. After 3 hours of incubation, tube-like structures were starting to form,

and firm tubes appeared from 4 to 6 hours after seeding the cells; over that time,

proteases released by cells started to disintegrate the existing tubes. So, in our

experiments, HUVECs were fixed and stained five hours after plating them in the

matrix. HUVECs treated with C5a showed more numerous tube-like structures than

cells in basal medium (Figure 26A). Analysis of the photographs taken in the central

area of wells demonstrated that total tube length was superior in cells treated with C5a

than in control cells (p=0.002) (Figure 26B).

Con

trol

C5a

0

1000

2000

3000

Tube length

(µµ µµm

/ field

)

**

A

B

Control C5a

Con

trol

C5a

0

1000

2000

3000

Tube length

(µµ µµm

/ field

)

**

A

B

Control C5a

Figure 26. C5a promotes tube formation by endothelial cells. HUVECs were plated on gelled Matrigel in the absence or presence of C5a at 10 ng/ml. After 5 hours, cells were fixed and stained. Three high-power fields per culture condition were examined using the 10X objective. A) Representative pictures are shown. B) For quantification of tube formation, the total length of each picture was measured using ImageJ software (NIH). Mean ± SEM is represented. Results are representative of three independent experiments, each one with at least five wells per condition. **p<0.01.

Page 124: Facultad de Ciencias - CORE

Results

95

4.4. Decreased tumor angiogenesis in C3 deficient animals

Due to our in vitro results demonstrating a role of C5a in the promotion of

angiogenesis, together with the in vivo impairment of tumor growth in animals deficient

in complement, we decided to analyze the effect of complement deficiency on the

promotion of angiogenesis. For this attempt, LLC cells were injected i.v. in the tail vein

of animals with normal (wild-type) or deficient complement (C3 knockout). Their lungs

were removed three weeks later and embedded in paraffin. The number of macroscopic

and microscopic (in H/E preparations) metastases were countered. No significant

differences between groups were found (data not shown). However the aspect of tumors

was qualitatively different between animals with normal complement and animals with

C3-deficiency. In the former, tumors seemed more hemorrhagic (data not shown).

CD31 immunostaining in lung slides showed a clear difference in microvessel density.

Tumors from C3-deficient mice presented a lower number of vessels than those from

wild-type mice (Figure 27A). The percentage of CD31 stained area was significantly

higher in tumors from wild-type mice than in those from C3-deficient animals

(p<0.001) (Figure 27B).

B

WT

C3 KO

0

2

4

6

8

Imm

unosta

ined a

rea (%

)

∗∗∗

AWT C3 KO

B

WT

C3 KO

0

2

4

6

8

Imm

unosta

ined a

rea (%

)

∗∗∗

WT

C3 KO

0

2

4

6

8

Imm

unosta

ined a

rea (%

)

∗∗∗

AWT C3 KO

Figure 27. Microvascular density, determined by CD31 immunohistochemistry, in lung tumors from wild type (WT) and C3 knockout (C3 KO) mice. A) A representative preparation from a tumor of each group is shown. B) Quantification of the microvascular density was carried out by measuring the fraction of immunostained area on tumor sections. Data show mean ± SEM. ***p < 0.001.

Page 125: Facultad de Ciencias - CORE

Results

96

4.5. C5aR-1 blockade and C3 deficiency reduces bFGF expression in the tumor

microenvironment

The expression of angiogenic factors was also studied in lung tumors from wild-

type animals treated with the C5aR-1 antagonist, the control peptide, or C3-deficient

animals treated with the control peptide. For this purpose, total RNA was extracted from

tumors and the expression of the angiogenic factors bFGF and VEGF was analyzed by

real time PCR. In the case of bFGF, tumors from wild-type animals treated with the

C5aR-1 antagonist showed a significantly lower expression of the factor (p.=0.015), and

this difference was even more pronounced in tumors from C3-deficient animals

(p=0.004) (Figure 28A). In the case of VEGF, the expression within the tumor

microenvironment was similar in all groups (Figure 28B).

∗∗

ns

ns

A B

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.00

0.02

0.04

0.06

bFG

F m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 ant

agon

ist

C3 KO + con

trol pep

tide

0.00

0.01

0.02

0.03

VEG

F m

RN

A (

ββ ββ-a

ctin %

)

∗∗

∗∗

ns

ns

A B

WT + co

ntro

l pep

tide

WT + C5a

R-1 antag

onist

C3 KO + con

trol pep

tide

0.00

0.02

0.04

0.06

bFG

F m

RN

A (

ββ ββ-a

ctin %

)

WT + co

ntro

l pep

tide

WT + C5a

R-1 ant

agon

ist

C3 KO + con

trol pep

tide

0.00

0.01

0.02

0.03

VEG

F m

RN

A (

ββ ββ-a

ctin %

)

Figure 28. Expression of the angiogenic factors bFGF and VEGF within the tumor microenvironment. Total mRNA was extracted from tumors of wild-type (WT) mice treated with the control peptide, wild-type mice treated with the C5aR-1 antagonist, and C3 knockout (KO) mice treated with the control peptide. Quantification of bFGF (A) and VEGF (B) was carried out by real-time PCR and the expression of each gene was calculated as percentage relative to the β-actin mRNA in each tumor. Individual results per animal and mean per group are shown. *p<0.05; **p<0.01.

Page 126: Facultad de Ciencias - CORE

Results

97

5. EFFECT OF C5a ON NON-SMALL CELL LUNG CANCER

CELLS

Once we had demonstrated the action of C5a on the regulation of immune

response and angiogenesis, our last question was whether C5a has a direct effect on

cancer cells.

5.1. Expression of C5aR-1 in lung cancer cell lines

First, we analyzed whether lung cancer cells express C5aR-1. By conventional

PCR, we demonstrated that many lung cancer cells express C5aR-1 (Figure 29). In

particular, the expression in small-cell lung cancer (SCLC) cells was very frequent, with

only one negative cell line (H446) out of eight cell lines analyzed. Among NSCLC

cells, most of them also expressed the receptor, although the levels were apparently

lower than in SCLC cells. Carcinoid cells also showed a high expression of the receptor.

Since our current study focused on the effects of complement on NSCLC cells, we

selected three cell lines with this histology for further studies. We chose A549 and

H157 cells as C5aR-1 expressing cells. H661, a cell line with undetected levels of the

protein, was selected as control. Expression of C5aR-1 on these NSCLC cell lines was

confirmed by flow cytometry (Figure 29). Protein levels on the cell membrane

correlated with mRNA levels.

Page 127: Facultad de Ciencias - CORE

Results

98

Fluorescence intensity

Ce

lln

um

ber

100 101 102 103 104

H661

20

00

100 101 102 103 104

H15720

00

100 101 102 103 104

A549

20

00

A

B

H 6

9

H 8

2

H 1

87

H 2

09

H 3

45

H 4

17

H 4

46

H 5

10

H 2

3

H 4

41

H 2

087

Cal

u3

H 1

648

H C

C82

7

H 3

58

A54

9

H 3

22

SCLC ADC BAC

H 6

9

H 8

2

H 1

87

H 2

09

H 3

45

H 4

17

H 4

46

H 5

10

H 2

3

H 4

41

H 2

087

Cal

u3

H 1

648

H C

C82

7

H 3

58

A54

9

H 3

22

SCLC ADC BAC

H52

0

H 1

703

2170

H 1

57

H 2

26

H T

B58

H 1

299

H 4

60

97TM

1

103H

H 6

61

H 7

27

H 7

20

SQ LC Carc.

H52

0

H 1

703

2170

H 1

57

H 2

26

H T

B58

H 1

299

H 4

60

97TM

1

103H

H 6

61

H 7

27

H 7

20

SQ LC Carc.

C5aR-1

GAPDH

C5aR-1

GAPDH

Fluorescence intensity

Ce

lln

um

ber

100 101 102 103 104

H661

20

00

100 101 102 103 104

H15720

00

100 101 102 103 104

A549

20

00

Fluorescence intensity

Ce

lln

um

ber

100 101 102 103 104

H661

20

00

100 101 102 103 104

H15720

00

100 101 102 103 104

A549

20

00

100 101 102 103 104

H661

20

00

100 101 102 103 104

H661

20

00

100 101 102 103 104

H15720

00

100 101 102 103 104

H157

100 101 102 103 104

H15720

00

100 101 102 103 104

A549

20

00

100 101 102 103 104

A549

20

00

A

B

H 6

9

H 8

2

H 1

87

H 2

09

H 3

45

H 4

17

H 4

46

H 5

10

H 2

3

H 4

41

H 2

087

Cal

u3

H 1

648

H C

C82

7

H 3

58

A54

9

H 3

22

SCLC ADC BAC

H 6

9

H 8

2

H 1

87

H 2

09

H 3

45

H 4

17

H 4

46

H 5

10

H 2

3

H 4

41

H 2

087

Cal

u3

H 1

648

H C

C82

7

H 3

58

A54

9

H 3

22

SCLC ADC BAC

H52

0

H 1

703

2170

H 1

57

H 2

26

H T

B58

H 1

299

H 4

60

97TM

1

103H

H 6

61

H 7

27

H 7

20

SQ LC Carc.

H52

0

H 1

703

2170

H 1

57

H 2

26

H T

B58

H 1

299

H 4

60

97TM

1

103H

H 6

61

H 7

27

H 7

20

SQ LC Carc.

C5aR-1

GAPDH

C5aR-1

GAPDH

Figure 29. Expression of C5aR-1 in lung cancer cells. A) Conventional PCR analysis of C5aR-1 in lung cancer cell lines. Detection of GAPDH mRNA was used to ensure equal loading and RNA integrity. SCLC: small cell lung cancer; ADC: adenocarcinoma; BAC: bronchioloalveolar carcinoma; SQ: squamous cell carcinoma; LC: large cell carcinoma; Carc: carcinoid. B) Detection of C5aR-1 by flow cytometry in some NSCLC cell lines. The expression of C5aR-1 was measured as the increase in intensity in the green channel (white peak). Incubation of cells with a control isotype antibody was used as a negative control (shaded peak).

5.2. Activation of the C5a signaling pathway on NSCLC cells

We next assessed whether C5a was able to activate C5aR-1 on the surface of

NSCLC cells. C5aR-1 is a member of the seven transmembrane G protein-coupled

receptor superfamily. Upon interaction with its ligand, C5aR-1 is activated and induces

calcium mobilization. Therefore, intracellular calcium after incubation with C5a was

analyzed in A549 and H157 cells. Granulocytes were used as a positive controls.

Treatment with high concentrations of C5a (2 µg/ml) increased calcium influx in

granulocytes, but did not produce any change in the calcium content in A549 or H157

cells. Of note, these cell lines showed very low basal levels of calcium compared to

granulocytes. After evaluating calcium basal levels in other NSCLC cells, we found that

H1299 cells, which also express C5aR-1, have higher basal levels of calcium than A549

and H157. In this cell line, high concentrations of C5a (2 µg/ml) enhanced calcium

influx (Figure 30A).

It has been reported that in some cell types, ERK1/2 is activated by C5a

stimulation, leading to NF-κB activation and translocation to the nucleus160. ERK

phosphorylation was analyzed in A549 cells by Western blotting after 30-minute

Page 128: Facultad de Ciencias - CORE

Results

99

incubation with 10 ng/ml C5a. An increased in ERK phosphorilation was detected

compared to the phosporylation level in cells treated with PBS (Figure 30B). The

translocation of NF-κB to the nucleus after C5aR-1 activation in A549 cells was also

studied. After 45 minutes of incubation with 10 ng/ml C5a, a clear translocation of NF-

κB to the nucleus was observed. This effect was blocked with the C5aR-1 antagonist,

but not with the control peptide (Figure 30C).

PBS C5a

- Phospho-p44 MAPK- Phospho-p42 MAPK

- p44 MAPK- p42 MAPK

B C Control C5a

C5a + C5aR-1 antagonist C5a + control peptide

A

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

250

200

150

100

50

500

400

300

200

100

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

250

200

150

100

50

500

400

300

200

100

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

250

200

150

100

50

250

200

150

100

50

500

400

300

200

100

PBS C5a

Granulocytes

A549

H157

H1299

Time (seconds)

Flu

ore

sce

nc

e in

ten

sit

y

PBS C5aPBS C5a

- Phospho-p44 MAPK- Phospho-p42 MAPK

- p44 MAPK- p42 MAPK

B C Control C5a

C5a + C5aR-1 antagonist C5a + control peptide

A

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

250

200

150

100

50

500

400

300

200

100

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

250

200

150

100

50

500

400

300

200

100

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

0 50 100 150 200 250 300 0 50 100 150 200 250 300

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

80

60

40

20

250

200

150

100

50

250

200

150

100

50

500

400

300

200

100

PBS C5a

Granulocytes

A549

H157

H1299

Time (seconds)

Flu

ore

sce

nc

e in

ten

sit

y

Figure 30. Activation of the C5a signaling pathway in lung cancer cells after C5a treatment. A) Calcium flux in granulocytes and NSCLC cell lines after stimulation with C5a. Fluo-4-loaded cells were initially analyzed by flow cytometry to establish calcium basal levels. After two minutes, either PBS (negative control) or C5a were added and increase in the intracellular calcium flux was recorded in real time over 150 s. B) Western blot analysis for phospho-p44/p42 MAPK and total p44/p42 MAPK was carried out in extracts of A549 cells after 30 minutes of treatment with 10 ng/ml C5a. C) Confocal images showing NF-κB translocation to the nucleus in cells treated with 10 ng/ml C5a, with or without the C5aR-1 antagonist or the control peptide. NF-κB staining is shown in red. DAPI (blue) was used to stain nuclei.

Page 129: Facultad de Ciencias - CORE

Results

100

5.3. Effect of C5a on NSCLC cell proliferation

The next aspect to be analyzed was the effect of C5a on cell proliferation using

the MTT assay. The two positive cell lines for C5aR-1, A549 and H157, and the

negative one, H661, were tested. We established two conditions for the assay: C5a at

different concentrations, ranging from 0.03 to 500 ng/ml, in RPMI basal medium

(Figure 31A), or in RPMI medium with 0.5% FBS (Figure 31B). The last condition

was tested considering that cells may need a minimal quantity of serum, which would

not disturb the effect of C5a but benefit cell viability during the assay. In none of the

situations C5a affected cell proliferation.

contro

l

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.5

1.0

1.5

C5a (ng/ml)

Absorb

ance (540 n

m)

contro

l

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.1

0.2

0.3

0.4

0.5

C5a (ng/ml)

Absorb

ance (540 n

m)

contro

l

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.1

0.2

0.3

0.4

0.5

C5a (ng/ml)Absorb

ance (540 n

m)

contro

l

FBS 10

%0.03

0.06

0.12

50.25 0.

5 5 50 500

0.0

0.2

0.4

0.6

0.8

1.0

1.2

0.5% FBS + C5a (ng/ml)

Absorb

ance (540 n

m)

contro

l

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.2

0.4

0.6

0.8

1.0

0.5% FBS + C5a (ng/ml)

Absorb

ance (540 n

m)

cont

rol

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.2

0.4

0.6

0.8

1.0

0.5% FBS + C5a (ng/ml)

Absorb

ance (540 n

m)

A549 H157 H661A

B

contro

l

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.5

1.0

1.5

C5a (ng/ml)

Absorb

ance (540 n

m)

contro

l

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.1

0.2

0.3

0.4

0.5

C5a (ng/ml)

Absorb

ance (540 n

m)

contro

l

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.1

0.2

0.3

0.4

0.5

C5a (ng/ml)Absorb

ance (540 n

m)

contro

l

FBS 10

%0.03

0.06

0.12

50.25 0.

5 5 50 500

0.0

0.2

0.4

0.6

0.8

1.0

1.2

0.5% FBS + C5a (ng/ml)

Absorb

ance (540 n

m)

contro

l

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.2

0.4

0.6

0.8

1.0

0.5% FBS + C5a (ng/ml)

Absorb

ance (540 n

m)

cont

rol

FBS 10

%0.03

0,06

0,12

50,25 0,

5 5 50 500

0.0

0.2

0.4

0.6

0.8

1.0

0.5% FBS + C5a (ng/ml)

Absorb

ance (540 n

m)

A549 H157 H661A

B

Figure 31. Effect of C5a on NSCLC cell proliferation. Cells were incubated with C5a at different concentrations ranging from 0.03 to 500 ng/ml, in the absence (A) or presence (B) of 0.5% FBS. Positive controls were performed with 10% FBS. The proliferative response was evaluated using MTT assay. One representative experiment is shown in each case, and results are expressed as the mean absorbance ± SEM.

The absence of any effect on cell proliferation was confirmed by the analysis of

the cell cycle in cells treated with C5a. For this purpose, A549 cells were incubated with

10 ng/ml C5a or RPMI basal medium for 48 hours, and the cell cycle distribution was

assessed. No differences were seen between the two treatments (Figure 32A).

Finally, anchorage-independent growth of A549 cells was studied after treatment

with C5a. As a common malignant cell line, A549 cells have the ability to proliferate

Page 130: Facultad de Ciencias - CORE

Results

101

without attachment to a substratum. C5a did not modify this malignant feature of A549

cells (Figure 32B).

0

100

200

300

control

C5a

Co

lon

ies

/ w

ell

8 10 12

Days of incubation

Cell

nu

mb

er

Fluorescence intensity

200 400 600 800 1000

SubG0

SG0/G1

G2/M

0

100

2

00

3

00

40

0

500

A B

nsns ns

control C5aSub G0/G1 0.7% 0.63%

G0/G1 70.75% 67.92%

S 15.89% 19.52% G2/M 7.20% 6.36%

0

100

200

300

control

C5a

control

C5a

Co

lon

ies

/ w

ell

8 10 12

Days of incubation

Cell

nu

mb

er

Fluorescence intensity

200 400 600 800 1000

Cell

nu

mb

er

Fluorescence intensity

200 400 600 800 1000

SubG0

SG0/G1

G2/M

0

100

2

00

3

00

40

0

500

A B

nsnsnsns nsns

control C5aSub G0/G1 0.7% 0.63%

G0/G1 70.75% 67.92%

S 15.89% 19.52% G2/M 7.20% 6.36%

Figure 32. Cell cycle distribution and anchorage-independent growth of A549 cells incubated with C5a. Cells were treated 48 hours with or without 10 ng/ml C5a in basal medium. A) Cell cycle profile of control cells is represented in grey, and cell cycle profile of cells treated with C5a is depicted with a black line. B) After treatment with C5a, one thousand A549 cells were plated in soft agar with C5a at 10 ng/ml. Control cells were incubated in the absence of C5a. Experiments were performed in triplicate. The graph shows the number of colonies formed after 8, 10 and 12 days of incubation (mean ± SEM).

5.4. Effect of C5a in A549 migration

C5a is a potent chemoattractant that can stimulate monocytes/macrophages to

migrate to sites of wound healing and inflammation164. We studied the effect of C5a on

NSCLC cell migration.

The Boyden chamber assay was used to study the migration of A549 cells through

a microporous filter toward different concentrations of C5a placed under the filter. Cells

were allowed to migrate for 48 h to the other side. Afterwards, cells were fixed, stained,

and counted. C5a stimulated the migration of A549 cells when added within 0.5 and

50 ng/ml (p0.5=0.009; p5=0.004; p50=0.001) (Figure 33). It is interesting to note that the

addition of C5a at a high concentration (500 ng/ml) had the inverse effect (p<0.001).

Page 131: Facultad de Ciencias - CORE

Results

102

cont

rol

0.25 0.

5 5 50 500

0

2

4

6

8

10

C5a (ng/ml)

Cell

num

ber

/ field

Control 5 ng/ml C5a

A B

∗∗

∗∗ ∗∗

∗∗∗

cont

rol

0.25 0.

5 5 50 500

0

2

4

6

8

10

C5a (ng/ml)

Cell

num

ber

/ field

Control 5 ng/ml C5aControl 5 ng/ml C5a

A B

∗∗

∗∗ ∗∗

∗∗∗

Figure 33. Effect of C5a on A549 migration in the transwell assay. C5a was added at different concentrations in basal medium (0.25, 0.5, 5, 50, and 500 ng/ml) on the lower side of the transwell, while A549 cells were plated in the upper chamber. After 48 hours of incubation, cells that had migrated to the underside of the transwell were fixed and stained with crystal violet. A) Cell migration in each transwell was analyzed by counting the cells in eight different fields of the transwell (at 10x magnification) and calculating the mean value. Data are collected from three independent experiments and shown as mean ± SEM. **p<0.01; ***p<0.001. B) Representative images at x4 magnification of control cells and cells treated with C5a added at 5 ng/ml.

5.5. Effect of C5a in NSCLC cell adhesion to extracellular matrix proteins

To better characterize the mechanism by which NSCLC cells increase its

migration ability in the presence of C5a, we examined the role of C5a in the capacity of

A549, H157 and H661 cells to adhere to different extracellular matrix (ECM) proteins.

A549 cells showed high adhesion to collagens I and IV, intermediate adhesion to

fibronectin and laminin, and low adhesion to vitronectin and gelatin (Figure 34A).

Adhesion to collagen I was reduced when cells were treated with C5a at 10 ng/ml for 48

hours (p=0.007). This effect was inhibited in the presence of the C5aR-1 antagonist. A

more accurate analysis was done by giving to the basal adhesion a 100% value (Figure

34B). With this analysis, the differences between treatments became more significant. A

clear decrease in the adhesion to collagens and fibronectin was observed (pcol.I=0.001;

pcol.IV=0.002; pfib.<0.001). In the case of collagens, the C5aR-1 antagonist blocked the

effect, but not in the case of fibronectin (p=0.002).

The H157 cell line showed a similar behavior in the adhesion to ECM proteins,

but these cells presented higher adhesion to fibronectin than to collagens (Figure 34A).

Adhesion to collagen I and fibronectin was reduced in cells treated with C5a

(pcol.I<0.001; pfib.=0.043). C5aR-1 antagonist blocked the effect in the case of

fibronectin, but not in the case of collagen I (p=0.002). Using the basal adhesion level

as reference (100%), we observed a significant decrease in cell adhesion to collagen I

(p<0.001), which was not inhibited with the antagonist (p=0.014) (Figure 34B). There

Page 132: Facultad de Ciencias - CORE

Results

103

was also a decrease in the adhesion to fibronectin and to vitronectin (pfib.=0.006;

pvit.=0.013), which was blocked by the presence of the antagonist.

Finally, the negative cell line, H661, showed a different pattern of adhesion to

ECM proteins. These cells presented low adhesion to collagens and laminin, but high

adhesion to gelatin and fibronectin, and intermediate adhesion to vitronectin. As

expected, adhesion to those ECM proteins was not affected by C5a treatment (Figure

34a and 34B).

Collage

n I

Collage

n IV

Fibr

onec

tin

Vitron

ectin

Laminin

Gelatin

0

20

40

60

80

Adhesio

n to E

CM

pro

tein

s (%

)

Collage

n I

Collage

n IV

Fibr

onec

tin

Vitron

ectin

Laminin

Gelatin

0

20

40

60

80Adhesio

n to E

CM

pro

tein

s (%

)

Collage

n I

Collage

n IV

Fibr

onec

tin

Vitron

ectin

Laminin

Gelatin-10

20

50

80

Adhesio

n to E

CM

pro

tein

s (%

)

Collage

n I

Collage

n IV

Fibr

onec

tin

0

50

100

150

Adhesio

n to E

CM

pro

tein

s v

s. contr

ol cells

(%

)

Collage

n I

Collage

n IV

Fibr

onec

tin

Vitron

ectin

0

50

100

150

Adhesio

n to E

CM

pro

tein

s v

s. contr

ol cells

(%

)

Fibr

onec

tin

Gelatin

0

50

100

150

200

250

Adhesio

n to E

CM

pro

tein

s v

s. contr

ol cells

(%

)

Control C5a C5a + C5aR-1 antagnist

A549 H157 H661A

B

∗∗

∗∗∗

∗∗

∗∗

∗∗

∗∗∗∗∗

∗∗∗

∗∗ ∗∗

Collage

n I

Collage

n IV

Fibr

onec

tin

Vitron

ectin

Laminin

Gelatin

0

20

40

60

80

Adhesio

n to E

CM

pro

tein

s (%

)

Collage

n I

Collage

n IV

Fibr

onec

tin

Vitron

ectin

Laminin

Gelatin

0

20

40

60

80Adhesio

n to E

CM

pro

tein

s (%

)

Collage

n I

Collage

n IV

Fibr

onec

tin

Vitron

ectin

Laminin

Gelatin-10

20

50

80

Adhesio

n to E

CM

pro

tein

s (%

)

Collage

n I

Collage

n IV

Fibr

onec

tin

0

50

100

150

Adhesio

n to E

CM

pro

tein

s v

s. contr

ol cells

(%

)

Collage

n I

Collage

n IV

Fibr

onec

tin

Vitron

ectin

0

50

100

150

Adhesio

n to E

CM

pro

tein

s v

s. contr

ol cells

(%

)

Fibr

onec

tin

Gelatin

0

50

100

150

200

250

Adhesio

n to E

CM

pro

tein

s v

s. contr

ol cells

(%

)

Control C5a C5a + C5aR-1 antagnist

A549 H157 H661A

B

∗∗

∗∗∗

∗∗

∗∗

∗∗

∗∗∗∗∗

∗∗∗

∗∗ ∗∗

Figure 34. Effect of C5a on NSCLC adhesion to different ECM proteins. NSCLC cell lines A549, H157 and H661 were incubated with basal medium alone (control), 10 ng/ml C5a, or C5a and the inhibitor peptide for C5aR1 at 100 ng/ml. After 48 hours, cells were collected, loaded with calcein, and plated in 96 well plates, where the different ECM proteins were set: collagen I, collagen IV, fibronectin, vitronectin, laminin and gelatin. After 30 minutes, non-adherent cells were removed and fluorescence was measured. The percentage of cells adhered to each ECM protein was calculated using the fluorescence of the wells containing the total number of cell loaded into them (A). The percentage of adhesion respect to the basal conditions is also shown (B). Data are presented as mean ± SEM. *p<0.05; **p<0.01; ***p<0.001.

5.5.1 Mechanisms involved in the regulation of cell adhesion to ECM proteins

by C5a

5.4.1. Regulation of integrin expression.

Due to the fact that integrins are heterodimeric transmembrane glycoproteins that

mediate attachment between a cell and the ECM, we studied the possible effect of C5a

Page 133: Facultad de Ciencias - CORE

Results

104

on integrin expression on the cell membrane. The expression of several well-

characterized integrins, α and β subunits (α1, α4, αv, β1 and β3), and three

hetorodimers (α4β1, α5β1, αvβ3), was apparently unaffected by C5a treatment (Figure

35).

100 101 102 103 104

β1

100 101 102 103 104

αv

100 101 102 103 104

α4β1 (VLA4)

100 101 102 103 104

αVβ3

100 101 102 103 104

α5β1

100 101 102 103 104100 101 102 103 104

α1 α4

100 101 102 103 104

β3

Fluorescence intensity

Cell

nu

mb

er

20

00

20

00

20

00

20

00

20

00

20

00

20

00

20

00

100 101 102 103 104

β1

100 101 102 103 104

β1

100 101 102 103 104

αv

100 101 102 103 104

αv

100 101 102 103 104

α4β1 (VLA4)

100 101 102 103 104

α4β1 (VLA4)

100 101 102 103 104

αVβ3

100 101 102 103 104

αVβ3

100 101 102 103 104

α5β1

100 101 102 103 104

α5β1

100 101 102 103 104100 101 102 103 104

α1 α4

100 101 102 103 104100 101 102 103 104

α1 α4

100 101 102 103 104

β3

100 101 102 103 104

β3

Fluorescence intensity

Cell

nu

mb

er

20

00

20

00

20

00

20

00

20

00

20

00

20

00

20

00

Figure 35. Expression of different integrins in A549 cells after incubation with C5a. Cells were incubated 48 hours with or without 10 ng/ml C5a in basal medium. Expression of integrins in treated (dotted line) or untreated cells (black line) was assessed by flow cytometry. An isotype control antibody was used as a negative control (shaded peak).

5.5.2 Regulation of metalloprotease activity

The lower attachment of C5a treated cells to the extracellular matrix proteins may

be explained by the production of proteases that degrade those proteins. Therefore, we

analyze the metalloproteinase activity within the conditioned medium of cells treated

with 10 ng/ml C5a for 48 hours. We measured the kinetics of hydrolysis of two

synthetic fluorogenic peptides which contain specific amino acid sequences for different

metalloproteases. In particular, one of the peptides, named here as Substrate 1, is a

substrate for MMP-3 (stromelysin 1) and MMP-10 (stromelysin 2). The other

fluorogenic peptide, named here as Substrate 2, is recognized by MMP-1 (collagenase

1), MMP-2 (gelatinase A), MMP-7 (matrilysin), MMP-8 (collagenase 2), MMP-9

(gelatinase B), MMP-12 (macrophage elastase), MMP-13 (collagenase 3), MMP-14

(MT1-MMP), MMP-15 (MT2-MMP) and MMP-16 (MT3-MMP).

We observed that the conditioned medium from C5a-treated A549 cells presented

a higher protease activity against Substrate 1 than the conditioned medium from control

cells or from cells treated with C5a and the C5aR-1 antagonist (Figure 36A). The

increase in the slope in the linear phase of the fluorescence curve was used to quantify

the difference in the protease activity. The medium from A549 cells treated with C5a

showed 3.43 ± 1.5 times more activity than the control medium. In addition, in the case

of cells treated with C5a and the C5aR-1 antagonist, the ratio between slopes was 0.66 ±

Page 134: Facultad de Ciencias - CORE

Results

105

0.64, confirming that the increase in protease activity could be blocked with the

antagonist.

When Substrate 2 was used, the conditioned medium of C5a-treated cells showed

also a higher protease activity than the control medium, with a ratio of slopes of 2.59 ±

1.46 (Figure 36B). The medium from cells treated with C5a and the C5aR-1 antagonist

showed a similar protease activity than the control one. Of note, the linear phase of the

fluorescence curve was clearly delayed compared to that obtained with Substrate 1.

In the case of H157 cells, the conditioned medium from C5a treated-cells showed

more protease activity for Substrate 1 than the control medium. However, no differences

were found in the activity against Substrate 2 (data not shown).

Time (seconds)

Flu

ore

scen

ce in

ten

sit

y

BA

60

100

140

80

120

160

1000 2000 3000 4000 5000 6000 7000

C-

Ant.C5aR-1

170

250

330

410

490

210

290

370

450

1000 2000 3000 4000 5000 6000 7000

Ant.C5aR-1

C-

C5a C5a

Time (seconds)

Flu

ore

scen

ce in

ten

sit

y

BA

60

100

140

80

120

160

1000 2000 3000 4000 5000 6000 7000

C-

Ant.C5aR-1

60

100

140

80

120

160

1000 2000 3000 4000 5000 6000 7000

C-

Ant.C5aR-1

170

250

330

410

490

210

290

370

450

1000 2000 3000 4000 5000 6000 7000

Ant.C5aR-1

C-

C5a

170

250

330

410

490

210

290

370

450

1000 2000 3000 4000 5000 6000 7000

Ant.C5aR-1

C-

C5a C5a

Figure 36. Kinetic analysis of metalloproteinase-mediated hydrolysis of fluorogenic peptide substrates. Kinetic data for cleavage of Substrate 1, a fluorogenic peptide recognized by MMP-3 and -10 (A), or Substrate 2, recognized by MMP-1,-2,-7,-8,-9-12,-13,-14,-15 and -16 (B). Conditioned media from A549 cells incubated with basal medium (C-), 10 ng/ml C5a or C5a with 100 ng/ml C5aR-1 antagonist (Ant.C5aR-1) were used to perform the experiments. The reactions were allowed to proceed for 120 minutes, and fluorescence data were evaluated at 1-minute intervals. Fluorescence was quantified as arbitrary units, and progression curves were plotted for each reaction as a function of time. Data show representative experiments for each substrate.

Page 135: Facultad de Ciencias - CORE
Page 136: Facultad de Ciencias - CORE

DISCUSSION

Page 137: Facultad de Ciencias - CORE
Page 138: Facultad de Ciencias - CORE

Discussion

109

1. THE DUAL ROLE OF COMPLEMENT ACTIVATION

In this work, we have demonstrated that lung cancer cells are able to activate

complement spontaneously, leading to the deposition of complement proteins on the

cell membrane and to the release of complement components, such as C5a, to the

extracellular medium. The release of this anaphylatoxin is a crucial event that

contributes to tumor progression by the induction of inmunosuppression, angiogenesis

and tumor cell migration.

In the cancer setting, researchers have traditionally been focused on the

conventional role of complement for tagging, clearance and lysis of tumor cells; and,

consequently, have been working in strategies such as the development of antibody

mediated responses through complement dependent cytotoxicity (CDC)165. But our

results in the present work, in line with other recent findings, challenge the conventional

view of complement. It seems that elements of the complement system perversely assist

tumor growth; in particular, C5a anaphylatoxin, generated within the tumor tissue

through the activation of complement, seems to have a major role. During the course of

the present study, some other works have been published supporting the role of C5a on

tumor progression. C5a activity has been shown to be connected to the recruitment and

activation of myeloid-derived suppressor cells (MDSCs)107. MDSCs shield cancer cells

from T-cell-mediated immune attack166. C5a released on the tumor environment may

play a key role as a chemoattractant for a subpopulation of MDSCs morphologically

related to neutrophils (PMN-MDSCs) and as an activator for the production of reactive

oxygen species (ROS) and reactive nitrogen species (RNS) in the monocyte-like

subpopulation (MO-MDSCs). This effect on MDSCs is critical for their suppressive

capabilities on T-cell function167. In addition, inhibition of the C5aR-1 was found to be

as efficient in reducing tumor growth as the treatment with the well-recognized anti-

cancer chemotherapeutic paclitaxel107. Nevertheless, MDSCs may not be the only type

of cells involved in the immunosuppressive response to tumors. The present work

suggests that complement activation and C5a may influence the presence of different

populations of immune cells within the tumors, tilting the immune response to

immunosuppression.

Although the finding that complement elements can act as tumor promoters is

novel, the idea is entirely consistent with the growing recognition of the homeostatic

Page 139: Facultad de Ciencias - CORE

Discussion

110

function of the complement system. The uncovering of this new function of

complement connects with two prominent aspects of cancer research: first, the role of

the tumor microenvironment in the initiation, development and progression of

malignant lesions; second, the impact of the inflammatory response in cancer. The

debate about inflammation and cancer came up more than a century ago, with the

Virchow’s and Coley’s theories. While the former supported that chronic inflammation

can be a factor that increases the risk of cancer, the latter defended that acute

inflammation can result in the regression of malignant tumors. Both theories have been

sustained by a huge number of clinical examples. There is a growing body of evidence

that many malignancies are initiated by infections and the subsequent continuous

inflammatory reaction168. In the contrary, the use of Coley’s toxins was the only known

systemic treatment for cancer until 1940s, and represents a pioneering method to exploit

the host response against tumors169. Although both theories would seem contradictory,

the recent progress in tumor immunology put both of them in the same context. While

Virchow defined a long lasting inflammation, that has been demonstrated to encourage

the genesis and spread of tumors, Coley observed a brisk inflammation that could fight

cancer with some grade of success. The paradigm of how complement fits in this puzzle

is now easily understood considering the kind of inflammatory response that is

involved. As a component of the innate immunity, the activation of complement

generates some of the most powerful proinflammatory molecules in the body, including

most notably the anaphylatoxins C3a and C5a. Therefore, the tumor-promoting

properties of the complement system are consistent with the currently accepted view of

inflammation as exacerbating the growth of various cancers and the recognition of C5a

as a key regulator of the inflammatory response160. Conversely, an anticancer function

for complement is well recognized, for example in its contribution to the clinical

efficacy of monoclonal antibodies for the treatment of neoplasias170-171.

Page 140: Facultad de Ciencias - CORE

Discussion

111

2. TUMOR CELLS ACTIVATE AND EFFICIENTLY REGULATE

THE COMPLEMENT CASCADE

2.1. NSCLC cell lines spontaneously activate the complement system

Tumorigenesis is a multistep process in which normal cells acquire progressively

genetic and epigenetic alterations. This progression is analogous to Darwinian

evolution, in which the succession of genetic changes, each conferring one or another

type of essential alterations in cell physiology, leads to the progressive conversion of

normal human cells into cancer cells172. During the evolution of a cancer, the sequential

alterations produce changes in cell morphology and result in the expression of many

neoantigens that can be represented by a change in protein glycosylation or in the

expression and structure of membrane-bound proteins. These new elements that

distinguish cancer cells from their normal counterparts may well be recognized by the

immune system109. The complement system, as part of the innate immunity, can

recognize those changes and be activated in the suitable surfaces that bear the tumor-

associated antigens. According to this theory, clinical studies have suggested that the

complement system is activated in patients with cancer in response to the expression of

tumor-associated antigens173. Deposited complement proteins have been observed on

the surface of many human tumors, and elevations in complement levels or activity in

the sera of patients with neoplastic diseases have been described. Therefore, these

observations indicate that complement is activated in the tumor tissue or in its vicinity.

In the present work we have performed for the first time a comparative study of

the in vitro activation of the complement system in tumor and normal cells. In

particular, we have compared non-small cell lung cancer (NSCLC) cells and normal

bronchial epithelial cells. The spontaneous activation of complement was higher on

tumor cells than on normal cells. This result is in agreement with the immune

surveillance theory, which supports that the immune system surveys the body for tumor-

associated antigens for the elimination of tumors119. This motivating result opens more

questions than answers; for example, which are those tumor antigens that trigger the

spontaneous activation of the complement system? In the case of the alternative

pathway, it has been shown that C3b attaches preferably sugars174 and amino acid

hydroxyl groups (Thr, Ser and Tyr)175-176. Then, the activation is more efficient on some

surfaces depending on their polysaccharide and protein composition. Another

Page 141: Facultad de Ciencias - CORE

Discussion

112

mechanism that triggers the alternative pathway, uncovered in the recent years, is the

direct ability of properdin to initiate complement activation in specific surfaces

associated with infection or injury, acting as a pattern recognition molecule177-178. This

suggests that properdin may become localized on the altered surface of tumor cells.

There is evidence for the binding of properdin to mouse tumors previously treated with

photodynamic therapy (PDT). In those mice, the injection of properdin increased the

cure rates of PDT-treated tumors, suggesting that this protein can localize on the cell

surface and initiate autologous complement activation through the alternative

pathway179. However, till the date, there is not any study that demonstrates a selective

deposit of properdin on untreated tumor tissues.

Regarding our results, another possibility is the activation of the classical or the

lectin pathways by molecules on the surface of tumor cells which are able to bind and

activate C1q or MBL. To elucidate the possible activation of these two pathways, new

experiments using deficient serum in different complement factors, C1q, C2, C4, MBL

or factor B, should be performed. Nevertheless, the potential role of the classical or the

lectin pathways would not reject the importance of the alternative pathway, since this

pathway acts as a loop after the initial deposition of C3b, amplifying the activation of

the other two pathways. It has recently been shown that the alternative pathway might

account for up to 80–90% of total complement activation, even when this pathway is

not the one initially triggered180.

2.2. NSCLC cells regulate complement dependent cytotoxicity

In our experiments, the efficient and spontaneous activation of complement on

NSCLC cells did not lead to a noticeable effect in cell lysis. Although at first glance this

result appeared unexpected, it is well understood considering that the efficiency of

complement-mediated tumor cytotoxicity is hampered by various protective

mechanisms120. Most of the mechanisms to resist complement attack are also exploited

by normal cells to avoid accidental activation or bystander effects from a local

activation of complement. However, it is reasonable to think that cancer cells develop

additional mechanisms to avoid the harm caused by complement activation. In vitro

studies have shown that lung cancer cells are extremely resistant to CDC, and this

resistance is much higher than that observed in normal cells, such as human nasal

epithelium primary cell cultures134,144. One of the best understood protective mechanism

used by cancer cells is the over-expression of membrane-associated and/or soluble

Page 142: Facultad de Ciencias - CORE

Discussion

113

complement regulatory proteins. Many current hypotheses propose that expression of

these proteins on the neoplastic cell membrane protects tumors from complement

activation.

Several studies have analyzed the expression of complement regulators in primary

tumors and cancer cell lines. Membrane-bound complement regulatory proteins

(mCRPs) appear to be overexpressed on many tumor cell types. Most cancers express at

least two mCRPs, with the exception of CR1. However, it is surprising the huge

variation in mCRP expression that can be observed among tumor types and even among

different specimens of the same tumor type121,181. The evident consequence for a high

expression of one or more mCRPs by a tumor cell would be a higher resistance to

complement cytotoxicity. The expression of CD59 correlated with the resistance of

different types of malignant cell lines133,182-184. Regarding the role of CD55 and CD46 in

complement activation, an increased expression of those molecules has been reported in

many tumors185-191. The secretion of soluble forms of mCRPs can also regulate

complement activation in cancer patients112-113,120,184,192-196, although this mechanism is

not restricted to malignant diseases and can be also found under normal conditions65,197.

Based on our results, we can propose that NSCLC cells present efficient

mechanisms to resist complement activation due to two fundamental signs. First, the

insignificant cytotoxicity produced by the activation of the alternative pathway, even

when we observed a higher C3 deposition on tumor cells than on normal cells. Second,

the low increase in C3 deposition by the classical pathway compared to the alternative

one. These facts imply indirectly that tumor cells are equipped with potent mechanisms

to challenge strong complement activation on their surface. However, in the present

study no clear differences were detected among lung tumor and normal cells in terms of

the expression of inhibitors. It is possible that there is not a predominant mechanism for

the control of complement activation by lung cancer cells. In fact, although there are a

high number of studies suggesting that tumor cells increase the expression of inhibitors,

some of them also show no differences or a reduction in the expression of particular

complement inhibitory proteins. The loss of one complement inhibitory protein may be

compensated by an increased expression of one of the others194. Although not uniformly

observed in all cell lines, in some lung cancer cell lines, we detected high expression

levels of the soluble regulator complement factor H. The contribution of soluble

complement regulatory proteins in the control of complement activation on cancer cells

had been underestimated until some works highlighted that these factors have a key role

Page 143: Facultad de Ciencias - CORE

Discussion

114

in the resistance of tumor cells to complement activation and CDC. This is the case of

factor H and its splicing product factor H-like protein 1 (FHL-1), whose expressions

have been described in primary tumors and cell lines from different origins198-202. In

fact, a clinically approved immunoassay for the detection of bladder cancer in urine is

based in the quantification of factor H203. We have found that factor H is elevated in

bronchoalveolar lavage and sputum from lung cancer patients. This finding is doubly

interesting. On the one hand, reasserts factor H as an active inhibitory mechanism in

complement activation in lung cancer. On the other hand, it introduces factor H as a

molecular biomarkers that may enhance the sensitivity of cytologic examination in the

detection of lung cancer146. Moreover, complement factor H antibodies in serum have

recently been found in lung cancer patients and its use as a molecular marker for early-

stage disease has been proposed204.

The expression of factor H and FHL-1, and their subsequent binding to the cell

membrane, may have consequences in the resistance of cancer cells against

complement. H2 glioblastoma cells showed an exceptional resistance to CDC even after

the combined neutralization of CD46, CD55, and CD59, but the use of anti-factor H

monoclonal antibodies enhanced cell death, confirming that factor H (or FHL-1) was

involved in the complement resistance of this cancer cell line205. Similarly, an anti-

factor H antibody also enhanced complement-mediated killing of Raji cells, a cell line

obtained from a Burkitt's lymphoma206. In lung cancer cells, the neutralization of CD46

and CD59 using specific antibodies was not effective in increasing the susceptibility to

CDC134, whereas the same antibodies successfully facilitated CDC on normal

respiratory epithelial cells144. These findings suggested that lung cancer cells have other

mechanisms to resist complement activation in addition to CD46 and CD59. In this

sense, our group demonstrated that NSCLC commonly expressed and secreted factor H

and FHL-1 to the extracellular milieu, where they were able to bind to lung tumor cell

surfaces and inhibit efficiently the activation of complement122. In addition, factor H

expression was knockdown in A549 lung cell line, reducing its growth significantly when

injected into athymic mice123.

The inhibitor C4b-binding protein (C4BP), the functional analogue of factor H in the

classical pathway, may be also playing a key role in complement regulation on cancer cells.

In a collaborative study with Professor Anna Blom our laboratory reported that NSCLC

cells produce the soluble complement inhibitors factor I and C4BP, and these cells are able

Page 144: Facultad de Ciencias - CORE

Discussion

115

to bind C4BP to their surface and support the cleavage of C3b and C4b124. This issue may

enable NSCLC cells to turn off the complement cascade.

In a different scenario, using hypoxic conditions to mimic in vitro the

circumstances of growing tumors, we observed that H358 and H2087 cell lines, two

NSCLC cell lines that express factor H, factor I and mCRPs, presented substantial C3

and C9 deposition after 24 hours of hypoxia and 24 hours of reoxygenation, and even

more after 48 hours of hypoxia. Nevertheless, the considerable complement activation

on those cells was not sufficient to cause CDC. The existence of hypoxic areas is a

common feature that occurs within tumors. Its appearance can be explained by the rapid

consumption of oxygen due to the sudden expanding of tumor cells and the bad quality

of the new vessels generated. Tumor vessels are often blind ended, have incomplete

endothelial lining and basement membrane and have a tendency to collapse207.

Furthermore, sudden restoration of oxygen (reoxygenation) worsens hypoxic injury

through increased ROS production208. The activation of complement on cells

undergoing hypoxic stress or hypoxia/reoxygenation has been widely observed in

several pathologies 209, although the molecules that trigger the different pathways are

unknown. One possibility is the generation of molecular changes in proteins and lipid

membranes after reacting with ROS208, or the production of new carbohydrate patterns

by the activation of certain genes that control sugar metabolism210. Those antigens may

potentially activate the three complement pathways211. This is in agreement with our

results, because NSCLC cell lines exposed to hypoxia or hypoxia/reoxygenation

showed an increase in C3 and C9 deposition when the experimental conditions

promoted exclusively the activation of the alternative pathway (Veronal buffer with

Mg2+ and EGTA) or the classical and lectin pathways (factor B-depleted serum in buffer

with Mg2+ and Ca2+). The binding of immunoglobulins, present in serum, C1q or MBL

to the surface of hypoxic cells sustained that classical and lectin pathways were also

activated. By contrast, those tumor cells, that efficiently activated complement,

appeared to be really resistant to CDC, as no increase in cell lysis was observed. This

again suggests that inhibitory mechanisms stopped complement cascade to elicit its

dreadful effects. Another remarkable finding of this study was that NSCLC cell lines

downregulated the expression of factor H, factor I, CD46 and, in a lower extent, CD55,

both in hypoxia/reoxygenation and even more in hypoxia. This implies that under

hypoxic conditions NSCLC cells reduce their mechanisms to face complement attack

while keep free from CDC.

Page 145: Facultad de Ciencias - CORE

Discussion

116

3. EFFECT OF COMPLEMENT ACTIVATION IN THE TUMOR

MICROENVIRONMENT

3.1. Complement deficiency impairs tumor growth

In the last years, complement activation has been exploited in the monoclonal

antibody-based immunotherapy against tumors, due to the improvement in efficacy

when different cytotoxic mechanisms are combined. There are several examples of

chimerized and humanized mouse monoclonal antibodies containing the human IgG1

Fc-region that binds the heterooligomeric complex C1q and activates the classical

complement pathway170. However, little attention has been paid to the promoting effect

of complement in malignant tumors. Using a syngeneic mouse model of lung cancer, we

have demonstrated that deficiency of complement impairs normal tumor growth. In

particular, we have shown that the deficiency in C3 and the blockade of C5aR-1 are

associated with tumor growth delay. This result indicates that complement is involved

in mechanisms that benefit the development of malignant tumors. Similar results were

obtained in a syngeneic mouse model of cervical cancer107, whereas in a transgenic

mouse model of epidermal neoplasia the authors showed that mechanisms related to

inflammation and tumor progression were independent of C3 deficiency212. These

contradictory findings may be due to the different situations where the complement

contribution to malignancy was studied. We and Markiewski’s group studied

complement effects in cells from advanced invasive tumors, whereas in the transgenic

model of epidermia neoplasia only premalignant skin lesions were considered.

Complement activation generates pro-inflammatory polypeptides named

anaphylatoxins, C3a, C4a and C5a. Within the group, C5a and C3a are both well known

for their immunoregulatory functions, whilst C4a function as an anaphylatoxin has been

recently questioned because of the lack of its own receptor and specific biological

functions100. The inflammatory potential of anaphylatoxins C3a and C5a must link

complement activation with the progression of tumors, in the same way it has been

implicated in the pathogenesis of many inflammatory and immunological diseases such

as sepsis, acute respiratory distress syndrome, rheumatoid arthritis, glomerulonephritis,

multiple sclerosis, ischemia-reperfusion injury, and asthma160. The harmful roles of

complement may be mainly due to the release of these activated products. Although

anaphylatoxins are not necessarily the initiating factors in the inflammatory disorders,

Page 146: Facultad de Ciencias - CORE

Discussion

117

they appear to be responsible for promoting and perpetuating inflammatory reactions.

The cleavage of C3 precedes the generation of complement inflammatory mediators,

although a local C5a generation in the absence of C3 in some pathological conditions

has been demonstrated34,36. In C3 deficient mice, it is difficult to ascertain whether the

pro-tumorigenic effects of complement activation are due to C3 or its downstream

products. However, the pharmacological blockade of C5aR-1 uncovered the role of C5a

in tumor progression. In any case, the effect of tumor regression was superior using C3

deficient mice than C5aR-1 antagonist. There may be two explanations for this fact:

first, the use of two different ways to achieve complement disruption, genetic deficiency

versus pharmacological blockade; and second, the assumption that C3a might also have

a tumor-promoting effect. Regarding to the second explanation, both C3a and C5a have

been shown to exert key regulatory functions in and out the immune system, through

their binding to three cognate receptors, i.e. C3aR-1, C5aR-1 and the C5a receptor-like

2, C5L2. However, C5aR-1 signaling has been the most characterized. The study of

C5aR-1 in a wide variety of cell types and the pleitropic biologic effects originated from

C5a binding (summarized in Table 7) made this anaphylatoxin an attractive starting

point for the study of complement effects in the tumor microenvironment. Besides, we

observed that C5a levels were increased in plasma and bronchoalveolar lavage

specimens from patients with lung cancer; suggesting that C5a is generated extensively

within tumors and diffuses systemically. In the tumor microenvironment, C5a may play

a key role in promoting tumor-associated effects. But it is important to keep in mind

that the role of C5a does not exclude the effects of C3a in tumor progression, an aspect

that warrants future studies.

Page 147: Facultad de Ciencias - CORE

Table 7. C5aR-1 functions on myeloid and non-myeloid cells.

Loci Cell type Action of C5a Reference

Myeloid cells Neutrophils Expression of adhesion molecules Superoxide release Release of granule enzymes Release of cytokines Reduction of apoptosis

Guo et al.160

Eosinophils Chemotaxis Release of granule enzymes

Guo et al.160

Basolphils Release of granule enzymes Kurimoto et al.213 Monocytes Release of cytokines

Chemotaxis Guo et al.160

Macrophages Enhancement of phagocytosis Release of cytokines

Guo et al.160

Mast cells Chemotaxis Release of histamine Change to prothombotic phenotype

Guo et al.160

Dendritic cells Activation and allostimulation of T cells Peng et al.214

Lymphocytes T cells Co-stimulation and suppression of apoptosis Strainic et al.92

Tonsil B cells Chemotaxis Ottonello et al.215

Thymocytes Apoptosis Riedemann216

Circulatory system HUVEC Induction of tissue factor activity Regulation of cytokine expression Upregulation of adhesion molecules in response to inflammation

Ikeda et al.217 Monsinjon et al.94 Foreman et al.218

Human immortalized dermal microvascular endothelial cells

Chemotaxis Schraufstatter et al.219

Septic cardiomiocites Reduction of contractibility Niederbichler220

Cerebral microvascular endothelial cells

Alteration of iNOS, ROS, and actin reorganization Jacob et al.221

Mouse microvascular endothelial cells

C5a synergistically enhanced chemokine release from MDMEC following exposure of endothelial cells to IL-6, IFN-γ, or LPS

Laudes et al.222

Page 148: Facultad de Ciencias - CORE

Loci Cell type Action of C5a Reference

CNS Neurons Possible protection during local inflammatory processes Apoptosis; neurodegeneration

O'Barr et al.223 Farkas et al.224

Microglia, reactive astrocytes Possible role in activation and recruitment of astrocites and microglia (gliosis) Gasque et al.225

Cultured neural stem cells Possible function in mammalian cerebrogenesis and basal adult neurogenesis Rahpeymai et al.226

Oligodendrocytes

Possible role in oligodendrocytes functions: migration, proliferation, regeneration, elimination of oligodendrocytes

Nataf et al.227

Connective tissue Synoviocytes Production of TNF and possible role in joint inflammation process Yuan et al.228

Synovial mast cells

Release of histamine in mast cells from rheumatoid arthritis Production of the proinflammatory cytokine IL-17A, which is considered a crucial player in rheumatoid arthritis

Kiener et al.229 Hueber et al.230

Human articular chondrocytes Possible effect of the cartilage pathology in rheumatoid arthritis Onuma et al.231

Eye Retinal pigment epithelial cells

Increase in the secretion of VEGF from ARPE-19 cells (role in AMD) Expression of VEGF, leukocyte recruitment and CNV formation in a mouse model of laser injury Induction of IL-8 mRNA (role in inflammation)

Cortright et al.232 Nozaki et al.233 Fukuoka et al.234

Kidney Cultured human renal glomerular mesangial cells

Promotion cellular proliferation and production of cytokines (MCP-1) and growth factors (PDGF-AB)

Braun et al.235

Human renal proximal tubular cells Unknown function in normal kidney Fayyazi et al.236

Mouse tubular epithelial cells Overexpression of both PDGF-B and -D and TGF-beta-1, acting as a profibrotic factor

Boor et al.237

Liver Human hepatoma-derived cell line HepG2

Production of alpha 1-antitrypsin Regulation of the expression of acute phase proteins

Buchner et al.238 McCoy et al.239

Stimulated hepatocytes

Enhancement of glucose output Regeneration of rat hepatocytes

Koleva et al.240 Daveau et al.241

Hepatic stellate and Kupfer cells Upregulation of extracellular matrix proteins Schlaf et al.242

Lung Human and mouse bronchial epithelial and smooth muscle cells, alveolar epithelial cells, and pulmonary smooth muscle endothelial cells

Participation in the pathology of diseases such as sepsis and asthma

Drouin et al.243

Rat alveolar epithelial cells Release of inflammatory mediators Riedemann et al.244

Skin Inflamed keratinocytes

Possible role in the pathogenesis of skin disorders beyond its well-defined function as a chemoattractant and activator of leukocytes

Fayyazi et al.245

92,94,160,213-245

Page 149: Facultad de Ciencias - CORE

Discussion

120

3.2. Complement proteins and immunosuppression

In this work we have also demonstrated that both C3 deficiency and the

pharmacological blockade of C5aR-1 quantitatively alters the populations of

splenocytes in the syngeneic model of lung cancer. CD19+ and CD8+ populations were

increased in C3 deficient mice while myeloid-derived suppressor cells (MDSC) and

regulatory T cells (Tregs) were decreased in those animals as compared to wild-type

mice. The same tendency was observed after blocking C5aR-1, although significant

differences were only seen in total MDSC, and the two characterized subpopulations of

these cells. MDSCs are a heterogeneous population of myeloid progenitor cells and

immature myeloid cells. Their immunosuppressive function consists in reducing

activated T-cells and disrupting their effects by multiple mechanisms, such as depletion

of L-arginine by arginase-1 (ARG-1) and production of nitric oxide by inducible nitric

oxide synthase (iNOS), which leads to an increased production of reactive oxygen

species (ROS), including peroxynitrites (ONOO) and hydrogen peroxide (H2O2).

MDSCs lack the expression of cell-surface markers that are specifically expressed by

monocytes, macrophages or dendritic cells. In mice, MDSCs are characterized by the

co-expression of the myeloid-cell lineage differentiation antigen GR1 and CD11b. The

differential expression of two epitopes in GR1, Ly6G and Ly6C (encoded by different

genes), has revealed a morphological heterogeneity within MDSCs. The use of two

epitope-specific antibodies has led to the identification of two MDSC subsets:

granulocytic MDSCs have a CD11b+Ly6G+Ly6Clow phenotype, whereas monocytic

MDSCs are CD11b+Ly6G-Ly6Chigh. In summary, the Ly6G molecule is expressed

primarily on granulocytes161, whereas Ly6C is typically highly expressed on

monocytes162. In our experiments, we distinguished the two subpopulations using the

antibodies anti CD11b and anti-Ly6C, and assumed that within the CD11b+ cells those

with low and high expression of Ly6C corresponded to granulocytic and monocytic

subpopulations of MDSCs, respectively. The recognition of these two subsets is

important as each may have different functions in cancer and infectious and auto

immune diseases246. Both subsets of MDSC have different mechanisms of action to

suppress antigen-specific T-cell proliferation. In brief, granulocytic MDSCs express

high levels of ROS, and monocytic MDSCs express high levels of NO; both subsets

express ARG-1247. However it is not clear if both subsets differ in the suppression of T-

Page 150: Facultad de Ciencias - CORE

Discussion

121

cells. Apart from this, MDSCs have been implicated in a whole array of non-

immunological functions, such as the promotion of angiogenesis and metastasis248-249.

Besides cancer, an increased production of MDSCs has also been reported in a

number of pathological conditions including traumatic stress250, bacterial and parasitic

infections251, sepsis252 and transplantation253. In cancer, this population of cells

increases up to ten fold in the spleen of patients with different tumors; and in mice,

expansion of MDSC has been detected in practically all tumor models. However, the

extent of expansion varies between different tumor models (from 2 to 20 times), which

can be explained by the fact that MDSCs are produced in response to a variety of tumor-

derived factors247. We observed that the percentage of MDSCs increases approximately

three fold in the spleen of animals bearing tumors with normal complement activity,

which is in agreement with other studies that used the same syngeneic model of lung

cancer247. Then, C3 deficiency or C5aR-1 blockade disrupted this expansion, halving

the percentage of MDSCs. MDSCs, together with regulatory T cells (Tregs), also

decreased in the spleen of C3 deficient mice, promoting an immunosuppressive

environment in tumor-bearing hosts. Classically Tregs have been characterized by the

cellular markers CD4+CD25+FoxP3+, although there are other phenotypically distinct

regulatory T-cell populations that have been suggested, including CD8+ subsets254.

CD25, the IL-2 receptor, is essential for Tregs survival. Foxp3, the transcription factor

forkhead box P3, controls the genes encoding proteins capable of mediating Treg

suppressive function255. Tregs suppress both T cells and antigen presenting cells (APC)

by several proposed mechanisms that include production of IL-10, which can drastically

suppress APC and T cell function, production of TGF-β, which may suppress natural

killer cell function, competitive consumption of IL-2, which is a survival factor for

activated T cells, perforin- and granzyme-dependent killing of T cells and APCs,

CTLA-4 induction of indolamine 2,3-dioxygenase (IDO)-expressing APCs, which

suppress T cell activation and promote tolerance, and also, induction of PDL-1

expression on APCs, which renders them immunosuppressive256.

Foxp3+CD25+CD4+ Treg cells are produced in the thymus as a functionally

mature T-cell subpopulation, and constitute 5-10% of peripheral CD4+ cells in healthy

mice and humans257. But they accumulate in tumors due to the recruitment through the

chemokine ligand 22 (CCL22) produced by tumor cells and macrophages258, and self-

antigens that Tregs recognize on tumors259. Differentiation or expansion within the

tumor stroma and conversion from normal T cells have also be proposed260.

Page 151: Facultad de Ciencias - CORE

Discussion

122

Interestingly, we observed that the ratio between Tregs and total CD4+ was significantly

reduced in C3 deficient mice. Finally, CD8+ and CD19+ populations were increased

quantitatively in spleens of C3 deficient animals. This may indicate that complement

activation disrupts a cytotoxic response raised against tumors, as well as an immune

response mediated by B cells.

We also analyzed the immunosuppressive microenvironment within tumors.

Unfortunately, due to the wide heterogeneity of cell populations that made difficult to

select the desired immune cells, we were not able to draw clear conclusions from these

data. In order to check whether the analysis of splenocytes represented the tumor

microenvironment, the expression of different molecules was analyzed. Interestingly,

the expression of immunosuppressive molecules was significantly decreased in C3

deficient mice and mice treated with the C5aR-1 antagonist. Those molecules included:

IL-10, PDL-1, CTLA-4, LAG-3, ARG-1 and IL12p35. IL-10 is an important

immunomodulatory cytokine well known because of its anti-inflammatory properties261.

PDL-1 (programmed death ligand 1, also known as B7-H1) is constitutively expressed

in cells of the macrophage lineage and regulates cellular and humoral immune responses

through the PD-1 receptor on activated T and B cells. An aberrant PDL-1 expression,

described in a number of human malignancies, impairs both T cell function and

survival262. CTLA-4 (cytotoxic T-lymphocyte antigen 4, also known as CD152) inhibits

T-cell activation when binds to CD80 and CD86 on APCs263. Based on the results of a

phase III clinical trial, ipilimumab, a human monoclonal antibody against CTLA-4, will

likely be approved soon by the FDA for metastatic melanoma. According to a research

presented in December 2010 at the Chicago Multidisciplinary Symposium in Thoracic

Oncology, ipilimumab also showed benefit in advanced non-small cell lung cancer.

LAG-3 (lymphocyte activation gene 3, also known as CD223) is a CD4-related

activation-induced cell surface molecule that binds to MHC class II molecules with high

affinity and negatively regulates T cell expansion and homeostasis264. CTLA-4 and

LAG-3 expression by Tregs are involved in their immune-suppressive function265-266.

ARG-1, together with iNOS, takes part in the catabolism of L-arginine, a non-essential

amino acid necessary for T-cell proliferation. These two enzymes are expressed in

MDSCs and their action depletes L-arginine from the microenvironment, inhibiting T-

cell proliferation267. IL-12p35 is one of the subunits of the heterodimeric cytokine IL-12

and the more recently described IL-35; while IL-12 stimulates the growth and function

of T cells, IL-35 may be specifically produced by Tregs and is required for their

Page 152: Facultad de Ciencias - CORE

Discussion

123

suppressive activity268. The expression of IL-6 was also decreased in animals treated

with the C5aR-1 antagonist. IL-6 has been linked to the promotion of some types of

cancers due to its proinflammatory nature269. Regarding to the complement activation,

deficiency in C3a, C5a or blockade of C5aR-1 impairs the induction of IL-6 in

hepatocytes, which is necessary to prevent cell death270. Similar mechanisms may occur

in the tumor setting. Finally, CD70, a member of the TNF receptor superfamily,

decreased in tumors from C3-deficient mice. Upon binding to its ligand, CD27 sends

costimulatory signals for the expansion and maturation of antigen-specific T effector

cells, thus governing the magnitude and maintenance of the cytotoxic response. Its

normal expression is highly restricted, but is aberrantly expressed in hematologic

malignancies and solid tumors271.

Globally, considering the different populations of immune cells in spleens and the

cytokines within tumors in C3 deficient mice and mice treated with the C5aR-1

antagonist peptide, we can conclude that normal complement activation contributes to

an immunosuppressive microenvironment necessary for tumor growth.

3.3. Complement and angiogenesis

Angiogenesis is another key mechanism that contributes to tumor growth. At

present, the role of complement in angiogenesis is a matter of debate. There are studies

that support the pro-angiogenic potential of complement activation, whereas there are

others that sustain the anti-angiogenic properties of complement. Recently, the anti-

angiogenic effect of C3 and C5 was shown in a model of retinopathy of prematurity,

where C5a indirectly produced an anti-angiogenic effect by stimulating macrophages

toward an angiogenesis-inhibitory phenotype272. On the contrary, the role of

complement in the activation of angiogenesis has been demonstrated in age-related

macular degeneration (AMD), a disease caused by choroidal neovascularization (CNV).

Both C3a and C5a are present in the lipoproteinaceous deposits that appear between the

coroid and retinal pigmented epithelium (RPE), also called drusen, in patients with

AMD or animals with laser-induced CNV. Both anaphylatoxins seem to be involved in

the induction of VEGF expression in vitro and in vivo in RPE cells, thus promoting the

generation of new vessels233. In addition, MAC and C3 are deposited in the eyes of

animals submitted to laser-induced CNV, concomitantly with an increase in the

expression of the angiogenic factors VEGF, TGF-β2 and bFGF273.

Page 153: Facultad de Ciencias - CORE

Discussion

124

We investigated the extent of angiogenesis in tumor slices from wild-type and C3

deficient mice or from mice treated with either the C5aR antagonist or the control

peptide. The immunohistochemistry of CD31 showed only minimal differences among

groups that did not reach statistical significance. The same finding had been previously

observed in a mouse model of cervical cancer107. Nevertheless, using extracts from

those tumors, we observed that the expression of bFGF, a potent mitogen and

chemotactic factor for endothelial cells274, was significantly reduced in animals treated

with the C5aR-1 antagonist or in C3 deficient mice. This result points to a role of

complement activation in the angiogenesis of tumors, although the question of why

there were not differences in vascular density remains unclear. Two possible

explanations could justify this finding. First, angiogenesis is orchestrated by a balance

between a variety of activators and inhibitors275, so a deeper analysis of the different

anti- and pro-angiogenic factors within the tumors would be needed to clarify their

angiogenic potential. Second, complement activation may be important in the

promotion of angiogenesis in the early steps of tumor formation. To evaluate this

hypothesis we injected syngeneic lung cancer cells into the vein of wild-type and C3

deficient animals, and analyzed the vessels formed in the disseminated nodules three

weeks after injection. Interestingly, C3 deficient mice presented a significantly lower

vessel density within the lung micrometastases than wild-type animals, supporting the

role of complement in angiogenesis. In vitro studies, using human umbilical vein

endothelial cells (HUVECs), support this conclusion. The activation of C5aR-1 in

endothelial cells causes cell retraction, increased paracellular permeability and

eosinophil transmigration276. It has also been demonstrated that C5a induces genes that

promote the effects of VEGF277. With this in mind, we studied the effects of C5a on

HUVECs. Of note, we detected considerable levels of C5aR-1 by flow cytometry in

HUVECs. However, the mRNA levels, detected by real-time PCR, seemed very low

(data not shown). In fact, the expression of C5aR-1 in HUVECs is controversial278. In

any case, it would be incorrect to conclude that the activation of C5aR-1 in HUVECs is

functionally irrelevant based on its low levels of expression, because engagement of a

few hundred G protein-coupled receptors per cell is sufficient for maximal cell

activation279. Bearing this in mind, we investigated the effect of C5a in HUVEC

proliferation, migration and formation of tube-like structures in a gelled matrix. The

formation of tubes is a method widely utilized to assess compounds that either inhibit or

stimulate angiogenesis280. We did not observe any effect in proliferation after C5a

Page 154: Facultad de Ciencias - CORE

Discussion

125

treatment, although this effect was previously observed by other groups in HUVECs

and immortalized human microvascular endothelial cells281-282. On the other hand, C5a

induced migration of HUVECs and formation of tubes in gelled Matrigel. The

chemotactic property of C5a in HUVECs was also seen in previous studies219. However,

to our knowledge, this is the first work in which the capacity of C5a to induce the

formation of tube-like structures by endothelial cells is demonstrated. Taken together,

these facts strongly suggest that C5a has angiogenic properties.

4. EFFECT OF C5a ON TUMOR CELLS

In the present study we have analyzed for the first time the expression of C5aR-1

in different lung cancer cell lines. The expression of C5aR-1 in human lung epithelial

cells had previously been demonstrated283. Of note, small-cell lung cancer (SCLC) and

carcinoid cells presented higher levels of expression than NSCLC cells. The present

work focused on NSCLC cells, but the high expression in lung cancer cells of

neuroendocrine origin merits further studies.

C5a signaling through C5aR-1, a seven transmembrane G protein-coupled receptor

that signals through Gαi and G16, has been well characterized in both immune and non-

immune cells. ERK1/2, Akt, and JNK are activated in monocytes and neutrophils,

whereas macrophages activate MAPK p38 pathway in response to C5a. In endothelial

cells, C5a/C5aR-1 induces transactivation of the epidermal growth factor receptor,

enhancing their migration284. We observed that C5aR activation in A549 cells produced

ERK phosphorylation and NF-κB translocation to the nucleus, as it was previously

reported for monocytes160. In undifferentiated human neuroblastoma cells, NF-κB

activation after C5aR-1 stimulation produced neoplastic proliferation223. However, we

were unable to appreciate any difference in lung cancer cell proliferation after treatment

with C5a or the three anaphylatoxins combined. Although calcium influx has been

described for both C5a and C3a on several cell types285, preceding NF-κB translocation

to the nucleus286, we did not observe a calcium influx after treating A549 or H157 cells

with high doses of C5a. In H1299 cells, which have high levels of intracellular calcium,

C5a treatment increased calcium influx, suggesting that the basal levels of calcium are a

key factor for this effect.

Cancer cell migration and adhesion to extracellular matrix (ECM) proteins are

processes highly related to invasion and metastasis. The migration and establishment of

Page 155: Facultad de Ciencias - CORE

Discussion

126

neoplastic colonies, distant from their origin, is the culprit of around 90% of cancer

deaths287. Several types of proteins are involved in the process, but of great importance

are those which reorganize their surrounding microenvironment and permit cell

migration and invasion through the extracellular matrix and into distant tissues. Those

proteins include cell-to-cell adhesion proteins and proteases. C5a is well recognized as a

chemoattractant for leukocytes and endothelial cells, but we present here for the first

time the chemotactic effect of C5a on cancer cells. In addition, the significant reduction

in ECM adhesion after C5a treatment supports this capacity. In our attempt to uncover

the mechanism beyond this process, we investigated the effect of C5a in the expression

of several integrins that mediate the adhesion to different ECM proteins. Integrins are a

large family of heterodimeric transmembrane glycoproteins. They are implicated in cell

proliferation, migration and survival288. In humans, 18 α-subunits and 8 β-subunits can

form a total of 24 integrin heterodimers. For our analysis, we selected those involved

preferentially in the adhesion to collagen I, collagen IV and fibronectin, which were the

ECM proteins for which we observed significant differences. However, no changes in

the expression of these molecules were observed, suggesting that other mechanisms are

responsible for the changes in adhesion.

Alternative mechanisms that would explain the relationship between complement

and degradation of ECM proteins have previously been proposed. The degradation can

be achieved directly by complement proteins289 or by the activation of matrix

metalloproteinases (MMPs). For example, C1s was found to activate the zymogen of

MMP-9, a protease with established roles in the metastasis of cancers290. In general,

MMPs are a family of zinc-dependent endopeptidases categorized by their architectural

features. In humans, twenty three MMPs have been indentified. All of them share a

general structural blueprint with three common domains: the pro-peptide, the catalytic

domain, and the hemopexin-like C-terminal domain291. The interaction of a cysteine

residue of the pro-domain with the zinc ion of the catalytic site keeps MMP in an

enzymatically inactive state. To become proteolytically active, the disruption of this

interaction has to occur by chemical modification of the cysteine residue or proteolytic

removal of the pro-domain, catalyzed extracellularly by convertases such as plasmin or

other MMPs, or intracellularly by furin. Once activated, MMPs realize their functions.

The most known function is the degradation of the ECM proteins, but they are also

involved in complex mechanisms that modulate angiogenesis, signaling pathways and

the inflammatory response292. The function of MMPs in vivo depends on the local

Page 156: Facultad de Ciencias - CORE

Discussion

127

balance between them and their physiological inhibitors. The most important inhibitors

are the four tissue inhibitors of metalloproteinases (TIMPs)291.

Using specific fluorogenic peptides for MMPs, we observed that the supernatants

of cells treated with C5a held a higher proteolytic activity than those from cells treated

without the anaphylatoxin or with C5a and the antagonist of C5aR-1. The use of two

peptides with different specificity for MMPs allowed us to discern the possible MMPs

involved in the ECM cleavage. The cleavage of the specific peptide for MMP-3 and

MMP-10 (stromelysin 1 and 2, respectively) was more efficient than the cleavage of the

peptide recognized by MMP-1, -2, -7, -8, -9, -13, -14, -15, and -16. In our experiments,

we also used positive controls: recombinant MMP-10 and human serum. As expected,

MMP-10 efficiently cleaved the first peptide. However, MMP-10 also showed some

activity on the second peptide. This indicates that one specific protease can cleave its

substrate efficiently, but may also have some activity over other different substrates. By

extrapolating this fact to our results, we conclude that stromelysins may be the

metalloproteinases involved in the proteolysis of the ECM proteins once cells have been

treated with C5a. Further work is needed to confirm this point and to identify the

specific MMP involved in the process. In preliminar studies, the increase in MMP

activity seemed not to be regulated by gene expression, as no changes in the expression

of specific MMPs at the RNA level were observed (data not shown). Since MMP

function is regulated by the shift from the inactive to the active form and by the

inactivation by MMP inhibitors, the increase in MMP activation or the decrease in

MMP inhibition should be evaluated.

Page 157: Facultad de Ciencias - CORE

Discussion

128

5. COMPLEMENT IN HEALTH AND DISEASE

The present work offers additional support to the transition from the classical

perception of the human complement system to a wider perspective. That is to say, from

its role as a microbial defense system to a complex system that takes part on cell

homeostasis, immune surveillance and the sustention of disease when the balance

between activation-regulation becomes awry.

The immunosurveillance functions of complement and their biological results

have recently been reviewed by Ricklin et al.15, and are illustrated in Figure 37. The

spontaneous C3 activation (tick-over) on normal cell surfaces is well checked by the

normal presence of mCRPs and the recruitment of soluble inhibitors from their

surroundings; hence the silent controlled complement activation does not upset the

homeostasis in healthy tissues (Figure 37A). Nevertheless, there are different situations

where the complement activation and regulation becomes relevant. In the case of

microbial or foreign cells, complement activation triggered by different means (such as

patron recognition molecules (PRM) or antigen-antibody complexes), in the absence of

complement inhibitors, permits amplification of the cascade. As a result, complement

plays its classical role of eliminating intruders by opsonization, CDC and stimulation of

downstream immune responses triggered by complement degradation products (Figure

37B). In the case of apoptotic cells, the balance between complement activation and

regulation is extremely delicate. Complement is activated by PRMs bore on those

altered cells and, together with the simultaneous shedding of CD46 and CD59,

apoptotic cells become opsonized by C3b and C4b, and phagocyted. The presence of

residual mCRPs on their surface and the binding of soluble inhibitors prevent the

release of inflammatory effectors and the harmful result of complement mediated lysis

(Figure 37C). In the case of some inflammatory, autoimmune, neurodegenerative and

infectious diseases, the balance between complement activation and regulation is

disrupted. Globally, the cause of this imbalance is an inappropriate initiation of the

cascade or deficiencies in specific complement inhibitors. Several diseases have been

related to both situations. Systemic lupus erythematosus, rheumatoid arthritis,

isquemia/reperfusion injury and Alzheimer’s disease are few examples of an excessive

activation. Age-related macula degeneration, atypical hemolytic uremic syndrome,

hereditary angioedema and paroxistical nocturnal hemoglobinuria, are caused by

inadequate complement inhibition293. In such pathological conditions, complement

Page 158: Facultad de Ciencias - CORE

Discussion

129

sustains inflammation and perpetuates tissue damage. (Figure 37D). In recent years,

several reports have demonstrated that complement dysregulation in those diseases is

associated with mutations or polymorphisms in the components and regulators of the

alternative pathway C3 convertase294.

tick-over

sufficient

regulation

strong

activation

opsonization

amplification

danger

signaling

lysis

activation

sufficient

regulation

attenuated

signalingattenuated

lysis

clearance

activation

insufficient

regulation

lysis

proinflammatory

signaling

A B

C D

tick-over

sufficient

regulation

strong

activation

opsonization

amplification

danger

signaling

lysis

activation

sufficient

regulation

attenuated

signalingattenuated

lysis

clearance

activation

insufficient

regulation

lysis

proinflammatory

signaling

A B

C D

Figure 37. Drawing showing the immune surveillance functions of complement in different settings: healthy cells (A), foreign or microbial cells (B), apoptotic cells (C) and host cells in disease conditions (D). Based on Ricklin et al.15

At this point of the work, based on our results and the most recent literature, we

can propose a fifth biological situation for lung cancer cells, and most probably for

many other cancer types (Figure 38):

Page 159: Facultad de Ciencias - CORE

Discussion

130

- Cancer cells are recognized by complement due to the presence of tumor-

associated antigens on their surface, causing a substantial activation of the system in the

tumor microenvironment.

- Cancer cells regulate complement activation, preventing their elimination.

- Complement activated proteins, such as C5a, orchestrate several biological

processes that promote immunosuppression, angiogenesis and invasion.

activation

sufficient

regulation

proinflammatory

signaling

deficient

lysis

Invasion

Angiogenesis

Immunosuppression

activation

sufficient

regulation

proinflammatory

signaling

deficient

lysis

Invasion

Angiogenesis

Immunosuppression

Figure 38. Potential oncogenic roles for complement proteins in NSCLC cells.

Page 160: Facultad de Ciencias - CORE

CONCLUSIONS

Page 161: Facultad de Ciencias - CORE
Page 162: Facultad de Ciencias - CORE

Conclusions

133

1. Non-small cell lung cancer (NSCLC) cells activate complement spontaneously.

This activation is significantly higher than in normal bronchial epithelial cells, and may

be responsible for the high levels of C5a anaphylatoxin found in biological fluids from

NSCLC patients. However, acquisition of different inhibitory mechanisms by cancer

cells, such as factor H overexpression, makes them resistant to the harmful effects of

complement.

2. In a syngeneic mouse lung cancer model, complement activation favors tumor

progression. This tumor promoting-effect of complement is mediated at least in part by

C5aR-1.

3. Three major mechanisms may be involved in the tumor promoting-effects

mediated by C5aR-1:

3a. C5a, a potent chemoattractant for different immune cells, increases splenic

suppressor cell populations and immunosuppressive cytokines within the tumor.

3b. C5a promotes migration of endothelial cells and facilities angiogenesis.

3c. C5a reduces adhesion of NSCLC cells to extracellular matrix proteins and

enhances cell migration, possibly by the release of active metalloproteinases.

4. Overall, these results suggest that the complement system can perversely assist

lung cancer growth. This finding may be relevant in the development of complement-

mediated therapies against lung cancer.

Page 163: Facultad de Ciencias - CORE
Page 164: Facultad de Ciencias - CORE

REFERENCES

Page 165: Facultad de Ciencias - CORE
Page 166: Facultad de Ciencias - CORE

References

137

1 Jemal, A. et al. Cancer statistics, 2009. CA Cancer J Clin 59, 225-249 (2009).

2 Ferlay, J. et al. Estimates of the cancer incidence and mortality in Europe in

2006. Ann Oncol 18, 581-592 (2007).

3 Ginsberg, R. Lung Cancer. (Hamilton: BC Decker, 2002).

4 Zakowski, M. F. Pathology of small cell carcinoma of the lung. Semin Oncol 30,

3-8 (2003).

5 Detterbeck, F. C., Boffa, D. J. & Tanoue, L. T. The new lung cancer staging

system. Chest 136, 260-271 (2009).

6 Henschke, C. I. et al. Survival of patients with stage I lung cancer detected on

CT screening. N Engl J Med 355, 1763-1771 (2006).

7 Schrump, D. S., Altorki, N.K., Henschke, C.L., Carter, D., Turrisi, A.T.,&

Gutierrez, M.E. Cancer: Principles and practice of oncology 7th edn, 753–776

(Lippincott Williams and Wilkins, 2005).

8 Tockman, M. S., Anthonisen, N. R., Wright, E. C. & Donithan, M. G. Airways

obstruction and the risk for lung cancer. Ann Intern Med 106, 512-518 (1987).

9 de Torres, J. P. et al. Assessing the relationship between lung cancer risk and

emphysema detected on low-dose CT of the chest. Chest 132, 1932-1938

(2007).

10 Alberg, A. J., Ford, J. G. & Samet, J. M. Epidemiology of lung cancer: ACCP

evidence-based clinical practice guidelines (2nd edition). Chest 132, 29S-55S

(2007).

Page 167: Facultad de Ciencias - CORE

References

138

11 Amos, C. I. et al. Genome-wide association scan of tag SNPs identifies a

susceptibility locus for lung cancer at 15q25.1. Nat Genet 40, 616-622 (2008).

12 Thorgeirsson, T. E. et al. A variant associated with nicotine dependence, lung

cancer and peripheral arterial disease. Nature 452, 638-642 (2008).

13 McKay, J. D. et al. Lung cancer susceptibility locus at 5p15.33. Nat Genet 40,

1404-1406 (2008).

14 Walport, M. J. Complement. First of two parts. N Engl J Med 344, 1058-1066

(2001).

15 Ricklin, D., Hajishengallis, G., Yang, K. & Lambris, J. D. Complement: a key

system for immune surveillance and homeostasis. Nat Immunol 11, 785-797

(2010).

16 Law, S. K. A., & Reid, K.B.M. Complement. 2nd edn, (Oxford University

Press, 1995).

17 Nauta, A. J. et al. Direct binding of C1q to apoptotic cells and cell blebs induces

complement activation. Eur J Immunol 32, 1726-1736 (2002).

18 Sjoberg, A. P. et al. Short leucine-rich glycoproteins of the extracellular matrix

display diverse patterns of complement interaction and activation. Mol Immunol

46, 830-839 (2009).

19 McGrath, F. D. et al. Evidence that complement protein C1q interacts with C-

reactive protein through its globular head region. J Immunol 176, 2950-2957

(2006).

20 Ying, S. C., Gewurz, A. T., Jiang, H. & Gewurz, H. Human serum amyloid P

component oligomers bind and activate the classical complement pathway via

Page 168: Facultad de Ciencias - CORE

References

139

residues 14-26 and 76-92 of the A chain collagen-like region of C1q. J Immunol

150, 169-176 (1993).

21 Mitchell, D. A., Kirby, L., Paulin, S. M., Villiers, C. L. & Sim, R. B. Prion

protein activates and fixes complement directly via the classical pathway:

implications for the mechanism of scrapie agent propagation in lymphoid tissue.

Mol Immunol 44, 2997-3004 (2007).

22 Jiang, H., Cooper, B., Robey, F. A. & Gewurz, H. DNA binds and activates

complement via residues 14-26 of the human C1q A chain. J Biol Chem 267,

25597-25601 (1992).

23 Mayer, M. M. Mechanism of cytolysis by complement. Proc Natl Acad Sci U S

A 69, 2954-2958 (1972).

24 Muller-Eberhard, H. J. The membrane attack complex of complement. Annu Rev

Immunol 4, 503-528 (1986).

25 Thiel, S. et al. Interaction of C1q and mannan-binding lectin (MBL) with C1r,

C1s, MBL-associated serine proteases 1 and 2, and the MBL-associated protein

MAp19. J Immunol 165, 878-887 (2000).

26 Matsushita, M., Thiel, S., Jensenius, J. C., Terai, I. & Fujita, T. Proteolytic

activities of two types of mannose-binding lectin-associated serine protease. J

Immunol 165, 2637-2642 (2000).

27 Bexborn, F., Andersson, P. O., Chen, H., Nilsson, B. & Ekdahl, K. N. The tick-

over theory revisited: formation and regulation of the soluble alternative

complement C3 convertase (C3(H2O)Bb). Mol Immunol 45, 2370-2379 (2008).

28 Hourcade, D. E. Properdin and complement activation: a fresh perspective. Curr

Drug Targets 9, 158-164 (2008).

Page 169: Facultad de Ciencias - CORE

References

140

29 McMullen, M. E. et al. Mannose-binding lectin binds IgM to activate the lectin

complement pathway in vitro and in vivo. Immunobiology 211, 759-766 (2006).

30 Zhang, M. et al. Activation of the lectin pathway by natural IgM in a model of

ischemia/reperfusion injury. J Immunol 177, 4727-4734 (2006).

31 Wagner, E., Platt, J. L., Howell, D. N., Marsh, H. C., Jr. & Frank, M. M. IgG

and complement-mediated tissue damage in the absence of C2: evidence of a

functionally active C2-bypass pathway in a guinea pig model. J Immunol 163,

3549-3558 (1999).

32 Selander, B. et al. Mannan-binding lectin activates C3 and the alternative

complement pathway without involvement of C2. J Clin Invest 116, 1425-1434

(2006).

33 Markiewski, M. M., Nilsson, B., Ekdahl, K. N., Mollnes, T. E. & Lambris, J. D.

Complement and coagulation: strangers or partners in crime? Trends Immunol

28, 184-192 (2007).

34 Huber-Lang, M. et al. Generation of C5a in the absence of C3: a new

complement activation pathway. Nat Med 12, 682-687 (2006).

35 Maruo, K., Akaike, T., Ono, T., Okamoto, T. & Maeda, H. Generation of

anaphylatoxins through proteolytic processing of C3 and C5 by house dust mite

protease. J Allergy Clin Immunol 100, 253-260 (1997).

36 Huber-Lang, M. et al. Generation of C5a by phagocytic cells. Am J Pathol 161,

1849-1859 (2002).

37 Governa, M. et al. Variability of biological effects of silicas: different degrees of

activation of the fifth component of complement by amorphous silicas. Toxicol

Appl Pharmacol 208, 68-77 (2005).

Page 170: Facultad de Ciencias - CORE

References

141

38 Governa, M. et al. In vitro cleavage by asbestos fibers of the fifth component of

human complement through free-radical generation and kallikrein activation. J

Toxicol Environ Health A 59, 539-552 (2000).

39 Governa, M. et al. Cleavage of the fifth component of human complement and

release of a split product with C5a-like activity by crystalline silica through free

radical generation and kallikrein activation. Toxicol Appl Pharmacol 179, 129-

136 (2002).

40 Kirkitadze, M. D. & Barlow, P. N. Structure and flexibility of the multiple

domain proteins that regulate complement activation. Immunol Rev 180, 146-

161 (2001).

41 Beinrohr, L. et al. C1 inhibitor serpin domain structure reveals the likely

mechanism of heparin potentiation and conformational disease. J Biol Chem

282, 21100-21109 (2007).

42 Davis, A. E., 3rd, Mejia, P. & Lu, F. Biological activities of C1 inhibitor. Mol

Immunol 45, 4057-4063 (2008).

43 Blom, A. M., Villoutreix, B. O. & Dahlback, B. Complement inhibitor C4b-

binding protein-friend or foe in the innate immune system? Mol Immunol 40,

1333-1346 (2004).

44 Garcia de Frutos, P., Hardig, Y. & Dahlback, B. Serum amyloid P component

binding to C4b-binding protein. J Biol Chem 270, 26950-26955 (1995).

45 Zipfel, P. F., Jokiranta, T. S., Hellwage, J., Koistinen, V. & Meri, S. The factor

H protein family. Immunopharmacology 42, 53-60 (1999).

Page 171: Facultad de Ciencias - CORE

References

142

46 de Cordoba, S. R. & de Jorge, E. G. Translational mini-review series on

complement factor H: genetics and disease associations of human complement

factor H. Clin Exp Immunol 151, 1-13 (2008).

47 Edwards, A. O. et al. Complement factor H polymorphism and age-related

macular degeneration. Science 308, 421-424 (2005).

48 Hageman, G. S. et al. A common haplotype in the complement regulatory gene

factor H (HF1/CFH) predisposes individuals to age-related macular

degeneration. Proc Natl Acad Sci U S A 102, 7227-7232 (2005).

49 Haines, J. L. et al. Complement factor H variant increases the risk of age-related

macular degeneration. Science 308, 419-421 (2005).

50 Klein, R. J. et al. Complement factor H polymorphism in age-related macular

degeneration. Science 308, 385-389 (2005).

51 Nabil, K. et al. Identification of human complement factor H as a chemotactic

protein for monocytes. Biochem J 326 ( Pt 2), 377-383 (1997).

52 Iferroudjene, D., Schouft, M. T., Lemercier, C., Gilbert, D. & Fontaine, M.

Evidence for an active hydrophobic form of factor H that is able to induce

secretion of interleukin 1-beta or by human monocytes. Eur J Immunol 21, 967-

972 (1991).

53 Zipfel, P. F. & Skerka, C. Complement factor H and related proteins: an

expanding family of complement-regulatory proteins? Immunol Today 15, 121-

126 (1994).

54 Skerka, C. & Zipfel, P. F. Complement factor H related proteins in immune

diseases. Vaccine 26 Suppl 8, I9-14 (2008).

Page 172: Facultad de Ciencias - CORE

References

143

55 Podack, E. R. & Muller-Eberhard, H. J. SC5b-9 complex of complement:

formation of the dimeric membrane attack complex by removal of S-protein. J

Immunol 124, 1779-1783 (1980).

56 Schvartz, I., Seger, D. & Shaltiel, S. Vitronectin. Int J Biochem Cell Biol 31,

539-544 (1999).

57 Jenne, D. E. & Tschopp, J. Molecular structure and functional characterization

of a human complement cytolysis inhibitor found in blood and seminal plasma:

identity to sulfated glycoprotein 2, a constituent of rat testis fluid. Proc Natl

Acad Sci U S A 86, 7123-7127 (1989).

58 Falgarone, G. & Chiocchia, G. Chapter 8: Clusterin: A multifacet protein at the

crossroad of inflammation and autoimmunity. Adv Cancer Res 104, 139-170

(2009).

59 Fischer, E., Appay, M. D., Cook, J. & Kazatchkine, M. D. Characterization of

the human glomerular C3 receptor as the C3b/C4b complement type one (CR1)

receptor. J Immunol 136, 1373-1377 (1986).

60 Fearon, D. T. Regulation of the amplification C3 convertase of human

complement by an inhibitory protein isolated from human erythrocyte

membrane. Proc Natl Acad Sci U S A 76, 5867-5871 (1979).

61 Seya, T. & Atkinson, J. P. Functional properties of membrane cofactor protein

of complement. Biochem J 264, 581-588 (1989).

62 Marie, J. C. et al. Linking innate and acquired immunity: divergent role of CD46

cytoplasmic domains in T cell induced inflammation. Nat Immunol 3, 659-666

(2002).

Page 173: Facultad de Ciencias - CORE

References

144

63 Kemper, C. et al. Activation of human CD4+ cells with CD3 and CD46 induces

a T-regulatory cell 1 phenotype. Nature 421, 388-392 (2003).

64 Cattaneo, R. Four viruses, two bacteria, and one receptor: membrane cofactor

protein (CD46) as pathogens' magnet. J Virol 78, 4385-4388 (2004).

65 Medof, M. E., Walter, E. I., Rutgers, J. L., Knowles, D. M. & Nussenzweig, V.

Identification of the complement decay-accelerating factor (DAF) on epithelium

and glandular cells and in body fluids. J Exp Med 165, 848-864 (1987).

66 Medof, M. E., Kinoshita, T. & Nussenzweig, V. Inhibition of complement

activation on the surface of cells after incorporation of decay-accelerating factor

(DAF) into their membranes. J Exp Med 160, 1558-1578 (1984).

67 Abbott, R. J. et al. Structural and functional characterization of a novel T cell

receptor co-regulatory protein complex, CD97-CD55. J Biol Chem 282, 22023-

22032 (2007).

68 Meri, S. et al. Human protectin (CD59), an 18,000-20,000 MW complement

lysis restricting factor, inhibits C5b-8 catalysed insertion of C9 into lipid

bilayers. Immunology 71, 1-9 (1990).

69 Kimberley, F. C., Sivasankar, B. & Paul Morgan, B. Alternative roles for CD59.

Mol Immunol 44, 73-81 (2007).

70 Koski, C. L., Ramm, L. E., Hammer, C. H., Mayer, M. M. & Shin, M. L.

Cytolysis of nucleated cells by complement: cell death displays multi-hit

characteristics. Proc Natl Acad Sci U S A 80, 3816-3820 (1983).

71 Campbell, A. K., Daw, R. A., Hallett, M. B. & Luzio, J. P. Direct measurement

of the increase in intracellular free calcium ion concentration in response to the

action of complement. Biochem J 194, 551-560 (1981).

Page 174: Facultad de Ciencias - CORE

References

145

72 Morgan, B. P. & Walport, M. J. Complement deficiency and disease. Immunol

Today 12, 301-306 (1991).

73 Cole, D. S. & Morgan, B. P. Beyond lysis: how complement influences cell fate.

Clin Sci (Lond) 104, 455-466 (2003).

74 Davis, A. E., 3rd, Harrison, R. A. & Lachmann, P. J. Physiologic inactivation of

fluid phase C3b: isolation and structural analysis of C3c, C3d,g (alpha 2D), and

C3g. J Immunol 132, 1960-1966 (1984).

75 Klickstein, L. B., Barbashov, S. F., Liu, T., Jack, R. M. & Nicholson-Weller, A.

Complement receptor type 1 (CR1, CD35) is a receptor for C1q. Immunity 7,

345-355 (1997).

76 Ghiran, I. et al. Complement receptor 1/CD35 is a receptor for mannan-binding

lectin. J Exp Med 192, 1797-1808 (2000).

77 Taylor, R. P. et al. Immune complexes bound to the primate erythrocyte

complement receptor (CR1) via anti-CR1 mAbs are cleared simultaneously with

loss of CR1 in a concerted reaction in a rhesus monkey model. Clin Immunol

Immunopathol 82, 49-59 (1997).

78 Bacle, F., Haeffner-Cavaillon, N., Laude, M., Couturier, C. & Kazatchkine, M.

D. Induction of IL-1 release through stimulation of the C3b/C4b complement

receptor type one (CR1, CD35) on human monocytes. J Immunol 144, 147-152

(1990).

79 Krych-Goldberg, M. & Atkinson, J. P. Structure-function relationships of

complement receptor type 1. Immunol Rev 180, 112-122 (2001).

Page 175: Facultad de Ciencias - CORE

References

146

80 Fearon, D. T. & Carter, R. H. The CD19/CR2/TAPA-1 complex of B

lymphocytes: linking natural to acquired immunity. Annu Rev Immunol 13, 127-

149 (1995).

81 Dempsey, P. W., Allison, M. E., Akkaraju, S., Goodnow, C. C. & Fearon, D. T.

C3d of complement as a molecular adjuvant: bridging innate and acquired

immunity. Science 271, 348-350 (1996).

82 van Lookeren Campagne, M., Wiesmann, C. & Brown, E. J. Macrophage

complement receptors and pathogen clearance. Cell Microbiol 9, 2095-2102

(2007).

83 Helmy, K. Y. et al. CRIg: a macrophage complement receptor required for

phagocytosis of circulating pathogens. Cell 124, 915-927 (2006).

84 Gorski, J. P., Hugli, T. E. & Muller-Eberhard, H. J. Characterization of human

C4a anaphylatoxin. J Biol Chem 256, 2707-2711 (1981).

85 Lee, D. K. et al. Identification of four novel human G protein-coupled receptors

expressed in the brain. Brain Res Mol Brain Res 86, 13-22 (2001).

86 Samson, M. et al. ChemR23, a putative chemoattractant receptor, is expressed in

monocyte-derived dendritic cells and macrophages and is a coreceptor for SIV

and some primary HIV-1 strains. Eur J Immunol 28, 1689-1700 (1998).

87 Ames, R. S., Tornetta, M. A., Foley, J. J., Hugli, T. E. & Sarau, H. M. Evidence

that the receptor for C4a is distinct from the C3a receptor.

Immunopharmacology 38, 87-92 (1997).

88 Boulay, F., Mery, L., Tardif, M., Brouchon, L. & Vignais, P. Expression cloning

of a receptor for C5a anaphylatoxin on differentiated HL-60 cells. Biochemistry

30, 2993-2999 (1991).

Page 176: Facultad de Ciencias - CORE

References

147

89 Ohno, M. et al. A putative chemoattractant receptor, C5L2, is expressed in

granulocyte and immature dendritic cells, but not in mature dendritic cells. Mol

Immunol 37, 407-412 (2000).

90 Gerard, N. P. et al. An anti-inflammatory function for the complement

anaphylatoxin C5a-binding protein, C5L2. J Biol Chem 280, 39677-39680

(2005).

91 Chen, N. J. et al. C5L2 is critical for the biological activities of the

anaphylatoxins C5a and C3a. Nature 446, 203-207 (2007).

92 Strainic, M. G. et al. Locally produced complement fragments C5a and C3a

provide both costimulatory and survival signals to naive CD4+ T cells. Immunity

28, 425-435 (2008).

93 Gasque, P. et al. The receptor for complement anaphylatoxin C3a is expressed

by myeloid cells and nonmyeloid cells in inflamed human central nervous

system: analysis in multiple sclerosis and bacterial meningitis. J Immunol 160,

3543-3554 (1998).

94 Monsinjon, T. et al. Regulation by complement C3a and C5a anaphylatoxins of

cytokine production in human umbilical vein endothelial cells. FASEB J 17,

1003-1014 (2003).

95 Fregonese, L. et al. Expression of the anaphylatoxin receptors C3aR and C5aR

is increased in fatal asthma. J Allergy Clin Immunol 115, 1148-1154 (2005).

96 Werfel, T. et al. Activated human T lymphocytes express a functional C3a

receptor. J Immunol 165, 6599-6605 (2000).

Page 177: Facultad de Ciencias - CORE

References

148

97 Monk, P. N., Scola, A. M., Madala, P. & Fairlie, D. P. Function, structure and

therapeutic potential of complement C5a receptors. Br J Pharmacol 152, 429-

448 (2007).

98 Okinaga, S. et al. C5L2, a nonsignaling C5A binding protein. Biochemistry 42,

9406-9415 (2003).

99 Gao, H. et al. Evidence for a functional role of the second C5a receptor C5L2.

FASEB J 19, 1003-1005 (2005).

100 Haas, P. J. & van Strijp, J. Anaphylatoxins: their role in bacterial infection and

inflammation. Immunol Res 37, 161-175 (2007).

101 Strey, C. W. et al. The proinflammatory mediators C3a and C5a are essential for

liver regeneration. J Exp Med 198, 913-923 (2003).

102 Benard, M. et al. Characterization of C3a and C5a receptors in rat cerebellar

granule neurons during maturation. Neuroprotective effect of C5a against

apoptotic cell death. J Biol Chem 279, 43487-43496 (2004).

103 Matthews, K. W., Mueller-Ortiz, S. L. & Wetsel, R. A. Carboxypeptidase N: a

pleiotropic regulator of inflammation. Mol Immunol 40, 785-793 (2004).

104 Ember, J. A., Jagels, M.A. & Hugli, T.E. Characterization of complement

anaphylatoxins and their biological responses. 241–284 (Marcel Dekker,

1998).

105 Oppermann, M. & Gotze, O. Plasma clearance of the human C5a anaphylatoxin

by binding to leucocyte C5a receptors. Immunology 82, 516-521 (1994).

Page 178: Facultad de Ciencias - CORE

References

149

106 Scola, A. M., Johswich, K. O., Morgan, B. P., Klos, A. & Monk, P. N. The

human complement fragment receptor, C5L2, is a recycling decoy receptor. Mol

Immunol 46, 1149-1162 (2009).

107 Markiewski, M. M. et al. Modulation of the antitumor immune response by

complement. Nat Immunol 9, 1225-1235 (2008).

108 Hakomori, S. Tumor malignancy defined by aberrant glycosylation and

sphingo(glyco)lipid metabolism. Cancer Res 56, 5309-5318 (1996).

109 Pardoll, D. Does the immune system see tumors as foreign or self? Annu Rev

Immunol 21, 807-839 (2003).

110 Niculescu, F., Rus, H. G., Retegan, M. & Vlaicu, R. Persistent complement

activation on tumor cells in breast cancer. Am J Pathol 140, 1039-1043 (1992).

111 Yamakawa, M. et al. Protection of thyroid cancer cells by complement-

regulatory factors. Cancer 73, 2808-2817 (1994).

112 Niehans, G. A., Cherwitz, D. L., Staley, N. A., Knapp, D. J. & Dalmasso, A. P.

Human carcinomas variably express the complement inhibitory proteins CD46

(membrane cofactor protein), CD55 (decay-accelerating factor), and CD59

(protectin). Am J Pathol 149, 129-142 (1996).

113 Bjorge, L. et al. Ascitic complement system in ovarian cancer. Br J Cancer 92,

895-905 (2005).

114 Ytting, H., Jensenius, J. C., Christensen, I. J., Thiel, S. & Nielsen, H. J.

Increased activity of the mannan-binding lectin complement activation pathway

in patients with colorectal cancer. Scand J Gastroenterol 39, 674-679 (2004).

Page 179: Facultad de Ciencias - CORE

References

150

115 Carli, M. et al. Fluctuation of serum complement levels in children with

neuroblastoma. Cancer 43, 2399-2404 (1979).

116 Maness, P. F. & Orengo, A. Serum complement levels in patients with digestive

tract carcinomas and other neoplastic diseases. Oncology 34, 87-89 (1977).

117 Matsutani, M. et al. Cellular immunity and complement levels in hosts with

brain tumours. Neurosurg Rev 7, 29-35 (1984).

118 Mangano, A., Messina, L., Birgillito, S., Stivala, F. & Bernardini, A.

Complement and its fractions (C3-C4) pattern in subjects with neoplasia. J

Immunopharmacol 6, 147-162 (1984).

119 Smyth, M. J., Godfrey, D. I. & Trapani, J. A. A fresh look at tumor

immunosurveillance and immunotherapy. Nat Immunol 2, 293-299 (2001).

120 Jurianz, K. et al. Complement resistance of tumor cells: basal and induced

mechanisms. Mol Immunol 36, 929-939 (1999).

121 Fishelson, Z., Donin, N., Zell, S., Schultz, S. & Kirschfink, M. Obstacles to

cancer immunotherapy: expression of membrane complement regulatory

proteins (mCRPs) in tumors. Mol Immunol 40, 109-123 (2003).

122 Ajona, D. et al. Expression of complement factor H by lung cancer cells: effects

on the activation of the alternative pathway of complement. Cancer Res 64,

6310-6318 (2004).

123 Ajona, D., Hsu, Y. F., Corrales, L., Montuenga, L. M. & Pio, R. Down-

regulation of human complement factor H sensitizes non-small cell lung cancer

cells to complement attack and reduces in vivo tumor growth. J Immunol 178,

5991-5998 (2007).

Page 180: Facultad de Ciencias - CORE

References

151

124 Okroj, M., Hsu, Y. F., Ajona, D., Pio, R. & Blom, A. M. Non-small cell lung

cancer cells produce a functional set of complement factor I and its soluble

cofactors. Mol Immunol 45, 169-179 (2008).

125 Kirschfink, M., Blase, L., Engelmann, S. & Schwartz-Albiez, R. Secreted

chondroitin sulfate proteoglycan of human B cell lines binds to the complement

protein C1q and inhibits complex formation of C1. J Immunol 158, 1324-1331

(1997).

126 Ollert, M. W. et al. C3-cleaving membrane proteinase. A new complement

regulatory protein of human melanoma cells. J Immunol 144, 3862-3867 (1990).

127 Paas, Y., Bohana-Kashtan, O. & Fishelson, Z. Phosphorylation of the

complement component, C9, by an ecto-protein kinase of human leukemic cells.

Immunopharmacology 42, 175-185 (1999).

128 Bohana-Kashtan, O., Pinna, L. A. & Fishelson, Z. Extracellular phosphorylation

of C9 by protein kinase CK2 regulates complement-mediated lysis. Eur J

Immunol 35, 1939-1948 (2005).

129 Morgan, B. P. Complement membrane attack on nucleated cells: resistance,

recovery and non-lethal effects. Biochem J 264, 1-14 (1989).

130 Reiter, Y., Ciobotariu, A. & Fishelson, Z. Sublytic complement attack protects

tumor cells from lytic doses of antibody and complement. Eur J Immunol 22,

1207-1213 (1992).

131 Schrama, D., Reisfeld, R. A. & Becker, J. C. Antibody targeted drugs as cancer

therapeutics. Nat Rev Drug Discov 5, 147-159 (2006).

Page 181: Facultad de Ciencias - CORE

References

152

132 Blok, V. T., Gelderman, K. A., Tijsma, O. H., Daha, M. R. & Gorter, A.

Cytokines affect resistance of human renal tumour cells to complement-

mediated injury. Scand J Immunol 57, 591-599 (2003).

133 Brasoveanu, L. I. et al. Levels of cell membrane CD59 regulate the extent of

complement-mediated lysis of human melanoma cells. Lab Invest 74, 33-42

(1996).

134 Varsano, S., Rashkovsky, L., Shapiro, H., Ophir, D. & Mark-Bentankur, T.

Human lung cancer cell lines express cell membrane complement inhibitory

proteins and are extremely resistant to complement-mediated lysis; a comparison

with normal human respiratory epithelium in vitro, and an insight into

mechanism(s) of resistance. Clin Exp Immunol 113, 173-182 (1998).

135 Paganelli, G. et al. Three-step monoclonal antibody tumor targeting in

carcinoembryonic antigen-positive patients. Cancer Res 51, 5960-5966 (1991).

136 Macor, P. et al. Complement activated by chimeric anti-folate receptor

antibodies is an efficient effector system to control ovarian carcinoma. Cancer

Res 66, 3876-3883 (2006).

137 Gelderman, K. A., Lam, S. & Gorter, A. Inhibiting complement regulators in

cancer immunotherapy with bispecific mAbs. Expert Opin Biol Ther 5, 1593-

1601 (2005).

138 Balkwill, F. & Mantovani, A. Inflammation and cancer: back to Virchow?

Lancet 357, 539-545 (2001).

139 Mantovani, A., Allavena, P., Sica, A. & Balkwill, F. Cancer-related

inflammation. Nature 454, 436-444 (2008).

Page 182: Facultad de Ciencias - CORE

References

153

140 Langman, M. J., Cheng, K. K., Gilman, E. A. & Lancashire, R. J. Effect of anti-

inflammatory drugs on overall risk of common cancer: case-control study in

general practice research database. BMJ 320, 1642-1646 (2000).

141 Colotta, F., Allavena, P., Sica, A., Garlanda, C. & Mantovani, A. Cancer-related

inflammation, the seventh hallmark of cancer: links to genetic instability.

Carcinogenesis 30, 1073-1081 (2009).

142 Laufer, J., Katz, Y. & Passwell, J. H. Extrahepatic synthesis of complement

proteins in inflammation. Mol Immunol 38, 221-229 (2001).

143 Nishioka, K., Kawamura, K., Hirayama, T., Kawashima, T. & Shimada, K. The

complement system in tumor immunity: significance of elevated levels of

complement in tumor bearing hosts. Ann N Y Acad Sci 276, 303-315 (1976).

144 Varsano, S., Frolkis, I., Shapiro, H. & Ophir, D. Human nasal epithelium

adsorbs complement C3-related fragments and expresses cell membrane

complement regulatory proteins. Laryngoscope 106, 599-604 (1996).

145 Gorter, A. et al. Expression of CD46, CD55, and CD59 on renal tumor cell lines

and their role in preventing complement-mediated tumor cell lysis. Lab Invest

74, 1039-1049 (1996).

146 Pio, R. et al. Complement factor H is elevated in bronchoalveolar lavage fluid

and sputum from patients with lung cancer. Cancer Epidemiol Biomarkers Prev

19, 2665-2672 (2010).

147 Sato, M. et al. Multiple oncogenic changes (K-RAS(V12), p53 knockdown,

mutant EGFRs, p16 bypass, telomerase) are not sufficient to confer a full

malignant phenotype on human bronchial epithelial cells. Cancer Res 66, 2116-

2128 (2006).

Page 183: Facultad de Ciencias - CORE

References

154

148 Reddel, R. R. et al. Development of tumorigenicity in simian virus 40-

immortalized human bronchial epithelial cell lines. Cancer Res 53, 985-991

(1993).

149 Ramirez, R. D. et al. Immortalization of human bronchial epithelial cells in the

absence of viral oncoproteins. Cancer Res 64, 9027-9034 (2004).

150 Bertram, J. S. & Janik, P. Establishment of a cloned line of Lewis Lung

Carcinoma cells adapted to cell culture. Cancer Lett 11, 63-73 (1980).

151 Sim, R. B., Day, A. J., Moffatt, B. E. & Fontaine, M. Complement factor I and

cofactors in control of complement system convertase enzymes. Methods

Enzymol 223, 13-35 (1993).

152 Finch, A. M. et al. Low-molecular-weight peptidic and cyclic antagonists of the

receptor for the complement factor C5a. J Med Chem 42, 1965-1974 (1999).

153 Morgan, B. Complement Methods and Protocols. Vol. 150 (Humana Press,

2000).

154 Servais, G., Walmagh, J. & Duchateau, J. Simple quantitative haemolytic

microassay for determination of complement alternative pathway activation

(AP50). J Immunol Methods 140, 93-100 (1991).

155 Irigoyen, M. et al. Hypoxia alters the adhesive properties of lymphatic

endothelial cells. A transcriptional and functional study. Biochim Biophys Acta

1773, 880-890 (2007).

156 Fehrenbach, H. Animal models of pulmonary emphysema: a stereologist’s

perspective. European Respiratory Review 15 (2006).

Page 184: Facultad de Ciencias - CORE

References

155

157 Treumann, A., Lifely, M. R., Schneider, P. & Ferguson, M. A. Primary structure

of CD52. J Biol Chem 270, 6088-6099 (1995).

158 Okroj, M., Corrales, L., Stokowska, A., Pio, R. & Blom, A. M. Hypoxia

increases susceptibility of non-small cell lung cancer cells to complement attack.

Cancer Immunol Immunother 58, 1771-1780 (2009).

159 Kock, M. A., Hew, B. E., Bammert, H., Fritzinger, D. C. & Vogel, C. W.

Structure and function of recombinant cobra venom factor. J Biol Chem 279,

30836-30843 (2004).

160 Guo, R. F. & Ward, P. A. Role of C5a in inflammatory responses. Annu Rev

Immunol 23, 821-852 (2005).

161 Fleming, T. J., Fleming, M. L. & Malek, T. R. Selective expression of Ly-6G on

myeloid lineage cells in mouse bone marrow. RB6-8C5 mAb to granulocyte-

differentiation antigen (Gr-1) detects members of the Ly-6 family. J Immunol

151, 2399-2408 (1993).

162 Sunderkotter, C. et al. Subpopulations of mouse blood monocytes differ in

maturation stage and inflammatory response. J Immunol 172, 4410-4417 (2004).

163 Kubota, Y., Kleinman, H. K., Martin, G. R. & Lawley, T. J. Role of laminin and

basement membrane in the morphological differentiation of human endothelial

cells into capillary-like structures. J Cell Biol 107, 1589-1598 (1988).

164 Snyderman, R., Pike, M. C., McCarley, D. & Lang, L. Quantification of mouse

macrophage chemotaxis in vitro: role of C5 for the production of chemotactic

activity. Infect Immun 11, 488-492 (1975).

Page 185: Facultad de Ciencias - CORE

References

156

165 Clynes, R. A., Towers, T. L., Presta, L. G. & Ravetch, J. V. Inhibitory Fc

receptors modulate in vivo cytoxicity against tumor targets. Nat Med 6, 443-446

(2000).

166 Nagaraj, S. & Gabrilovich, D. I. Tumor escape mechanism governed by

myeloid-derived suppressor cells. Cancer Res 68, 2561-2563 (2008).

167 Gabrilovich, D. I. & Nagaraj, S. Myeloid-derived suppressor cells as regulators

of the immune system. Nat Rev Immunol 9, 162-174 (2009).

168 Coussens, L. M. & Werb, Z. Inflammation and cancer. Nature 420, 860-867

(2002).

169 Davis, I. D., Jefford, M., Parente, P. & Cebon, J. Rational approaches to human

cancer immunotherapy. J Leukoc Biol 73, 3-29 (2003).

170 Gelderman, K. A., Tomlinson, S., Ross, G. D. & Gorter, A. Complement

function in mAb-mediated cancer immunotherapy. Trends Immunol 25, 158-164

(2004).

171 Hsu, Y. F. et al. Complement activation mediates cetuximab inhibition of non-

small cell lung cancer tumor growth in vivo. Mol Cancer 9, 139 (2010).

172 Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57-70

(2000).

173 Guidi, L. et al. Immune complexes in solid tumours precipitable by 3.5%

polyethylene glycol: analysis of some nonspecific components. Diagn Clin

Immunol 5, 284-288 (1988).

Page 186: Facultad de Ciencias - CORE

References

157

174 Sahu, A., Kozel, T. R. & Pangburn, M. K. Specificity of the thioester-containing

reactive site of human C3 and its significance to complement activation.

Biochem J 302 ( Pt 2), 429-436 (1994).

175 Sahu, A. & Pangburn, M. K. Covalent attachment of human complement C3 to

IgG. Identification of the amino acid residue involved in ester linkage formation.

J Biol Chem 269, 28997-29002 (1994).

176 Sahu, A. & Pangburn, M. K. Tyrosine is a potential site for covalent attachment

of activated complement component C3. Mol Immunol 32, 711-716 (1995).

177 Spitzer, D., Mitchell, L. M., Atkinson, J. P. & Hourcade, D. E. Properdin can

initiate complement activation by binding specific target surfaces and providing

a platform for de novo convertase assembly. J Immunol 179, 2600-2608 (2007).

178 Kemper, C., Atkinson, J. P. & Hourcade, D. E. Properdin: emerging roles of a

pattern-recognition molecule. Annu Rev Immunol 28, 131-155 (2010).

179 Korbelik, M. & Cecic, I. Complement activation cascade and its regulation:

relevance for the response of solid tumors to photodynamic therapy. J

Photochem Photobiol B 93, 53-59 (2008).

180 Harboe, M. & Mollnes, T. E. The alternative complement pathway revisited. J

Cell Mol Med 12, 1074-1084 (2008).

181 Weichenthal, M., Siemann, U., Neuber, K. & Breitbart, E. W. Expression of

complement regulator proteins in primary and metastatic malignant melanoma. J

Cutan Pathol 26, 217-221 (1999).

182 Jarvis, G. A. et al. Expression and function of the complement membrane attack

complex inhibitor protectin (CD59) in human prostate cancer. Int J Cancer 71,

1049-1055 (1997).

Page 187: Facultad de Ciencias - CORE

References

158

183 Bjorge, L., Vedeler, C. A., Ulvestad, E. & Matre, R. Expression and function of

CD59 on colonic adenocarcinoma cells. Eur J Immunol 24, 1597-1603 (1994).

184 Donin, N. et al. Complement resistance of human carcinoma cells depends on

membrane regulatory proteins, protein kinases and sialic acid. Clin Exp Immunol

131, 254-263 (2003).

185 Loberg, R. D., Wojno, K. J., Day, L. L. & Pienta, K. J. Analysis of membrane-

bound complement regulatory proteins in prostate cancer. Urology 66, 1321-

1326 (2005).

186 Mustafa, T. et al. Expression of CD97 and CD55 in human medullary thyroid

carcinomas. Int J Oncol 24, 285-294 (2004).

187 Loberg, R. D., Day, L. L., Dunn, R., Kalikin, L. M. & Pienta, K. J. Inhibition of

decay-accelerating factor (CD55) attenuates prostate cancer growth and survival

in vivo. Neoplasia 8, 69-78 (2006).

188 Durrant, L. G. et al. Enhanced expression of the complement regulatory protein

CD55 predicts a poor prognosis in colorectal cancer patients. Cancer Immunol

Immunother 52, 638-642 (2003).

189 Madjd, Z. et al. Loss of CD55 is associated with aggressive breast tumors. Clin

Cancer Res 10, 2797-2803 (2004).

190 Madjd, Z. et al. Do poor-prognosis breast tumours express membrane cofactor

proteins (CD46)? Cancer Immunol Immunother 54, 149-156 (2005).

191 Rushmere, N. K. et al. Analysis of the level of mRNA expression of the

membrane regulators of complement, CD59, CD55 and CD46, in breast cancer.

Int J Cancer 108, 930-936 (2004).

Page 188: Facultad de Ciencias - CORE

References

159

192 Sadallah, S. et al. Soluble complement receptor 1 is increased in patients with

leukemia and after administration of granulocyte colony-stimulating factor. J

Leukoc Biol 65, 94-101 (1999).

193 Seya, T. et al. Purification and functional properties of soluble forms of

membrane cofactor protein (CD46) of complement: identification of forms

increased in cancer patients' sera. Int Immunol 7, 727-736 (1995).

194 Li, L., Spendlove, I., Morgan, J. & Durrant, L. G. CD55 is over-expressed in the

tumour environment. Br J Cancer 84, 80-86 (2001).

195 Morgan, J., Spendlove, I. & Durrant, L. G. The role of CD55 in protecting the

tumour environment from complement attack. Tissue Antigens 60, 213-223

(2002).

196 Hakulinen, J., Junnikkala, S., Sorsa, T. & Meri, S. Complement inhibitor

membrane cofactor protein (MCP; CD46) is constitutively shed from cancer cell

membranes in vesicles and converted by a metalloproteinase to a functionally

active soluble form. Eur J Immunol 34, 2620-2629 (2004).

197 Davies, A. et al. CD59, an LY-6-like protein expressed in human lymphoid

cells, regulates the action of the complement membrane attack complex on

homologous cells. J Exp Med 170, 637-654 (1989).

198 Gasque, P. et al. Expression of complement components of the alternative

pathway by glioma cell lines. J Immunol 149, 1381-1387 (1992).

199 Gasque, P., Thomas, A., Fontaine, M. & Morgan, B. P. Complement activation

on human neuroblastoma cell lines in vitro: route of activation and expression of

functional complement regulatory proteins. J Neuroimmunol 66, 29-40 (1996).

Page 189: Facultad de Ciencias - CORE

References

160

200 Junnikkala, S. et al. Secretion of soluble complement inhibitors factor H and

factor H-like protein (FHL-1) by ovarian tumour cells. Br J Cancer 87, 1119-

1127 (2002).

201 Katz, Y., Guterman, M. & Lahat, E. Regulation of synthesis of complement

proteins in HEp2 cells. Clin Immunol Immunopathol 67, 117-123 (1993).

202 Legoedec, J., Gasque, P., Jeanne, J. F. & Fontaine, M. Expression of the

complement alternative pathway by human myoblasts in vitro: biosynthesis of

C3, factor B, factor H and factor I. Eur J Immunol 25, 3460-3466 (1995).

203 Cheng, Z. Z. et al. Complement factor H as a marker for detection of bladder

cancer. Clin Chem 51, 856-863 (2005).

204 Amornsiripanitch, N. et al. Complement factor H autoantibodies are associated

with early stage NSCLC. Clin Cancer Res 16, 3226-3231 (2010).

205 Junnikkala, S. et al. Exceptional resistance of human H2 glioblastoma cells to

complement-mediated killing by expression and utilization of factor H and

factor H-like protein 1. J Immunol 164, 6075-6081 (2000).

206 Corey, M. J., Kinders, R. J., Brown, L. G. & Vessella, R. L. A very sensitive

coupled luminescent assay for cytotoxicity and complement-mediated lysis. J

Immunol Methods 207, 43-51 (1997).

207 Brown, J. M. & Giaccia, A. J. The unique physiology of solid tumors:

opportunities (and problems) for cancer therapy. Cancer Res 58, 1408-1416

(1998).

208 Li, C. & Jackson, R. M. Reactive species mechanisms of cellular hypoxia-

reoxygenation injury. Am J Physiol Cell Physiol 282, C227-241 (2002).

Page 190: Facultad de Ciencias - CORE

References

161

209 Arumugam, T. V., Shiels, I. A., Woodruff, T. M., Granger, D. N. & Taylor, S.

M. The role of the complement system in ischemia-reperfusion injury. Shock 21,

401-409 (2004).

210 Kannagi, R. Carbohydrate antigen sialyl Lewis a--its pathophysiological

significance and induction mechanism in cancer progression. Chang Gung Med

J 30, 189-209 (2007).

211 Carroll, M. C. & Holers, V. M. Innate autoimmunity. Adv Immunol 86, 137-157

(2005).

212 de Visser, K. E., Korets, L. V. & Coussens, L. M. Early neoplastic progression

is complement independent. Neoplasia 6, 768-776 (2004).

213 Kurimoto, Y., de Weck, A. L. & Dahinden, C. A. Interleukin 3-dependent

mediator release in basophils triggered by C5a. J Exp Med 170, 467-479 (1989).

214 Peng, Q. et al. Dendritic cell function in allostimulation is modulated by C5aR

signaling. J Immunol 183, 6058-6068 (2009).

215 Ottonello, L. et al. rC5a directs the in vitro migration of human memory and

naive tonsillar B lymphocytes: implications for B cell trafficking in secondary

lymphoid tissues. J Immunol 162, 6510-6517 (1999).

216 Riedemann, N. C. et al. C5a receptor and thymocyte apoptosis in sepsis. FASEB

J 16, 887-888 (2002).

217 Ikeda, K. et al. C5a induces tissue factor activity on endothelial cells. Thromb

Haemost 77, 394-398 (1997).

218 Foreman, K. E. et al. C5a-induced expression of P-selectin in endothelial cells. J

Clin Invest 94, 1147-1155 (1994).

Page 191: Facultad de Ciencias - CORE

References

162

219 Schraufstatter, I. U., Trieu, K., Sikora, L., Sriramarao, P. & DiScipio, R.

Complement c3a and c5a induce different signal transduction cascades in

endothelial cells. J Immunol 169, 2102-2110 (2002).

220 Niederbichler, A. D. et al. An essential role for complement C5a in the

pathogenesis of septic cardiac dysfunction. J Exp Med 203, 53-61 (2006).

221 Jacob, A. et al. C5a alters blood-brain barrier integrity in experimental lupus.

FASEB J 24, 1682-1688 (2010).

222 Laudes, I. J. et al. Expression and function of C5a receptor in mouse

microvascular endothelial cells. J Immunol 169, 5962-5970 (2002).

223 O'Barr, S. A. et al. Neuronal expression of a functional receptor for the C5a

complement activation fragment. J Immunol 166, 4154-4162 (2001).

224 Farkas, I. et al. Complement C5a receptor-mediated signaling may be involved

in neurodegeneration in Alzheimer's disease. J Immunol 170, 5764-5771 (2003).

225 Gasque, P., Singhrao, S. K., Neal, J. W., Gotze, O. & Morgan, B. P. Expression

of the receptor for complement C5a (CD88) is up-regulated on reactive

astrocytes, microglia, and endothelial cells in the inflamed human central

nervous system. Am J Pathol 150, 31-41 (1997).

226 Rahpeymai, Y. et al. Complement: a novel factor in basal and ischemia-induced

neurogenesis. EMBO J 25, 1364-1374 (2006).

227 Nataf, S., Levison, S. W. & Barnum, S. R. Expression of the anaphylatoxin C5a

receptor in the oligodendrocyte lineage. Brain Res 894, 321-326 (2001).

Page 192: Facultad de Ciencias - CORE

References

163

228 Yuan, G. et al. Expression of C5aR (CD88) of synoviocytes isolated from

patients with rheumatoid arthritis and osteoarthritis. Chin Med J (Engl) 116,

1408-1412 (2003).

229 Kiener, H. P. et al. Expression of the C5a receptor (CD88) on synovial mast

cells in patients with rheumatoid arthritis. Arthritis Rheum 41, 233-245 (1998).

230 Hueber, A. J. et al. Mast cells express IL-17A in rheumatoid arthritis synovium.

J Immunol 184, 3336-3340 (2010).

231 Onuma, H. et al. Expression of the anaphylatoxin receptor C5aR (CD88) by

human articular chondrocytes. Rheumatol Int 22, 52-55 (2002).

232 Cortright, D. N., Meade, R., Waters, S. M., Chenard, B. L. & Krause, J. E. C5a,

but not C3a, increases VEGF secretion in ARPE-19 human retinal pigment

epithelial cells. Curr Eye Res 34, 57-61 (2009).

233 Nozaki, M. et al. Drusen complement components C3a and C5a promote

choroidal neovascularization. Proc Natl Acad Sci U S A 103, 2328-2333 (2006).

234 Fukuoka, Y. & Medof, E. M. C5a receptor-mediated production of IL-8 by the

human retinal pigment epithelial cell line, ARPE-19. Curr Eye Res 23, 320-325

(2001).

235 Braun, M. & Davis, A. E., 3rd. Cultured human glomerular mesangial cells

express the C5a receptor. Kidney Int 54, 1542-1549 (1998).

236 Fayyazi, A. et al. The C5a receptor is expressed in normal renal proximal

tubular but not in normal pulmonary or hepatic epithelial cells. Immunology 99,

38-45 (2000).

Page 193: Facultad de Ciencias - CORE

References

164

237 Boor, P. et al. Complement C5 mediates experimental tubulointerstitial fibrosis.

J Am Soc Nephrol 18, 1508-1515 (2007).

238 Buchner, R. R., Hugli, T. E., Ember, J. A. & Morgan, E. L. Expression of

functional receptors for human C5a anaphylatoxin (CD88) on the human

hepatocellular carcinoma cell line HepG2. Stimulation of acute-phase protein-

specific mRNA and protein synthesis by human C5a anaphylatoxin. J Immunol

155, 308-315 (1995).

239 McCoy, R. et al. N-formylpeptide and complement C5a receptors are expressed

in liver cells and mediate hepatic acute phase gene regulation. J Exp Med 182,

207-217 (1995).

240 Koleva, M. et al. Induction of anaphylatoxin C5a receptors in rat hepatocytes by

lipopolysaccharide in vivo: mediation by interleukin-6 from Kupffer cells.

Gastroenterology 122, 697-708 (2002).

241 Daveau, M. et al. Expression of a functional C5a receptor in regenerating

hepatocytes and its involvement in a proliferative signaling pathway in rat. J

Immunol 173, 3418-3424 (2004).

242 Schlaf, G. et al. Upregulation of fibronectin but not of entactin, collagen IV and

smooth muscle actin by anaphylatoxin C5a in rat hepatic stellate cells. Histol

Histopathol 19, 1165-1174 (2004).

243 Drouin, S. M. et al. Expression of the complement anaphylatoxin C3a and C5a

receptors on bronchial epithelial and smooth muscle cells in models of sepsis

and asthma. J Immunol 166, 2025-2032 (2001).

244 Riedemann, N. C. et al. Expression and function of the C5a receptor in rat

alveolar epithelial cells. J Immunol 168, 1919-1925 (2002).

Page 194: Facultad de Ciencias - CORE

References

165

245 Fayyazi, A. et al. C5a receptor and interleukin-6 are expressed in tissue

macrophages and stimulated keratinocytes but not in pulmonary and intestinal

epithelial cells. Am J Pathol 154, 495-501 (1999).

246 Youn, J. I. & Gabrilovich, D. I. The biology of myeloid-derived suppressor

cells: the blessing and the curse of morphological and functional heterogeneity.

Eur J Immunol 40, 2969-2975 (2010).

247 Youn, J. I., Nagaraj, S., Collazo, M. & Gabrilovich, D. I. Subsets of myeloid-

derived suppressor cells in tumor-bearing mice. J Immunol 181, 5791-5802

(2008).

248 Priceman, S. J. et al. Targeting distinct tumor-infiltrating myeloid cells by

inhibiting CSF-1 receptor: combating tumor evasion of antiangiogenic therapy.

Blood 115, 1461-1471 (2010).

249 Yang, L. et al. Abrogation of TGF beta signaling in mammary carcinomas

recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell 13,

23-35 (2008).

250 Makarenkova, V. P., Bansal, V., Matta, B. M., Perez, L. A. & Ochoa, J. B.

CD11b+/Gr-1+ myeloid suppressor cells cause T cell dysfunction after traumatic

stress. J Immunol 176, 2085-2094 (2006).

251 Voisin, M. B., Buzoni-Gatel, D., Bout, D. & Velge-Roussel, F. Both expansion

of regulatory GR1+ CD11b+ myeloid cells and anergy of T lymphocytes

participate in hyporesponsiveness of the lung-associated immune system during

acute toxoplasmosis. Infect Immun 72, 5487-5492 (2004).

252 Delano, M. J. et al. MyD88-dependent expansion of an immature GR-

1(+)CD11b(+) population induces T cell suppression and Th2 polarization in

sepsis. J Exp Med 204, 1463-1474 (2007).

Page 195: Facultad de Ciencias - CORE

References

166

253 Suzuki, J., Ricordi, C. & Chen, Z. Immune tolerance induction by integrating

innate and adaptive immune regulators. Cell Transplant 19, 253-268 (2010).

254 Cosmi, L. et al. Human CD8+CD25+ thymocytes share phenotypic and

functional features with CD4+CD25+ regulatory thymocytes. Blood 102, 4107-

4114 (2003).

255 Gavin, M. A. et al. Foxp3-dependent programme of regulatory T-cell

differentiation. Nature 445, 771-775 (2007).

256 Cao, X. Regulatory T cells and immune tolerance to tumors. Immunol Res 46,

79-93 (2010).

257 Sakaguchi, S. Naturally arising Foxp3-expressing CD25+CD4+ regulatory T

cells in immunological tolerance to self and non-self. Nat Immunol 6, 345-352

(2005).

258 Curiel, T. J. et al. Specific recruitment of regulatory T cells in ovarian

carcinoma fosters immune privilege and predicts reduced survival. Nat Med 10,

942-949 (2004).

259 Nishikawa, H. et al. Definition of target antigens for naturally occurring CD4(+)

CD25(+) regulatory T cells. J Exp Med 201, 681-686 (2005).

260 Wilke, C. M., Wu, K., Zhao, E., Wang, G. & Zou, W. Prognostic significance of

regulatory T cells in tumor. Int J Cancer 127, 748-758 (2010).

261 Grutz, G. New insights into the molecular mechanism of interleukin-10-

mediated immunosuppression. J Leukoc Biol 77, 3-15 (2005).

262 Dong, H. et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential

mechanism of immune evasion. Nat Med 8, 793-800 (2002).

Page 196: Facultad de Ciencias - CORE

References

167

263 Scalapino, K. J. & Daikh, D. I. CTLA-4: a key regulatory point in the control of

autoimmune disease. Immunol Rev 223, 143-155 (2008).

264 Workman, C. J., Dugger, K. J. & Vignali, D. A. Cutting edge: molecular

analysis of the negative regulatory function of lymphocyte activation gene-3. J

Immunol 169, 5392-5395 (2002).

265 Read, S., Malmstrom, V. & Powrie, F. Cytotoxic T lymphocyte-associated

antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory

cells that control intestinal inflammation. J Exp Med 192, 295-302 (2000).

266 Huang, C. T. et al. Role of LAG-3 in regulatory T cells. Immunity 21, 503-513

(2004).

267 Bronte, V. & Zanovello, P. Regulation of immune responses by L-arginine

metabolism. Nat Rev Immunol 5, 641-654 (2005).

268 Collison, L. W. et al. The inhibitory cytokine IL-35 contributes to regulatory T-

cell function. Nature 450, 566-569 (2007).

269 Danese, S. & Mantovani, A. Inflammatory bowel disease and intestinal cancer: a

paradigm of the Yin-Yang interplay between inflammation and cancer.

Oncogene 29, 3313-3323 (2010).

270 Markiewski, M. M. et al. The regulation of liver cell survival by complement. J

Immunol 182, 5412-5418 (2009).

271 McEarchern, J. A. et al. Engineered anti-CD70 antibody with multiple effector

functions exhibits in vitro and in vivo antitumor activities. Blood 109, 1185-

1192 (2007).

Page 197: Facultad de Ciencias - CORE

References

168

272 Langer, H. F. et al. Complement-mediated inhibition of neovascularization

reveals a point of convergence between innate immunity and angiogenesis.

Blood 116, 4395-4403 (2010).

273 Bora, P. S. et al. Role of complement and complement membrane attack

complex in laser-induced choroidal neovascularization. J Immunol 174, 491-497

(2005).

274 Cross, M. J. & Claesson-Welsh, L. FGF and VEGF function in angiogenesis:

signalling pathways, biological responses and therapeutic inhibition. Trends

Pharmacol Sci 22, 201-207 (2001).

275 Bergers, G. & Benjamin, L. E. Tumorigenesis and the angiogenic switch. Nat

Rev Cancer 3, 401-410 (2003).

276 Jagels, M. A., Daffern, P. J. & Hugli, T. E. C3a and C5a enhance granulocyte

adhesion to endothelial and epithelial cell monolayers: epithelial and endothelial

priming is required for C3a-induced eosinophil adhesion. Immunopharmacology

46, 209-222 (2000).

277 Albrecht, E. A. et al. C5a-induced gene expression in human umbilical vein

endothelial cells. Am J Pathol 164, 849-859 (2004).

278 Zwirner, J., Fayyazi, A. & Gotze, O. Expression of the anaphylatoxin C5a

receptor in non-myeloid cells. Mol Immunol 36, 877-884 (1999).

279 Sklar, L. A. et al. Signal transduction and ligand-receptor dynamics in the

human neutrophil. Transient responses and occupancy-response relations at the

formyl peptide receptor. J Biol Chem 260, 11461-11467 (1985).

280 Ponce, M. L. Methods in Molecular Medicine. Vol. 46 (Humana Press Inc.,

2001).

Page 198: Facultad de Ciencias - CORE

References

169

281 Langeggen, H., Johnson, E. & Hetland, G. Effects of C5a and FMLP on

interleukin-8 production and proliferation of human umbilical vein endothelial

cells. Inflammation 25, 83-89 (2001).

282 Kurihara, R. et al. C5a promotes migration, proliferation, and vessel formation

in endothelial cells. Inflamm Res 59, 659-666 (2010).

283 Floreani, A. A. et al. Expression of receptors for C5a anaphylatoxin (CD88) on

human bronchial epithelial cells: enhancement of C5a-mediated release of IL-8

upon exposure to cigarette smoke. J Immunol 160, 5073-5081 (1998).

284 Rutkowski, M. J., Sughrue, M. E., Kane, A. J., Mills, S. A. & Parsa, A. T.

Cancer and the complement cascade. Mol Cancer Res 8, 1453-1465 (2010).

285 Monk, P. N., Barker, M. D. & Partridge, L. J. Multiple signalling pathways in

the C5a-induced expression of adhesion receptor Mac-1. Biochim Biophys Acta

1221, 323-329 (1994).

286 Crane, J. W. et al. The C5a anaphylatoxin receptor CD88 is expressed in

presynaptic terminals of hippocampal mossy fibres. J Neuroinflammation 6, 34

(2009).

287 Christofori, G. Changing neighbours, changing behaviour: cell adhesion

molecule-mediated signalling during tumour progression. EMBO J 22, 2318-

2323 (2003).

288 Desgrosellier, J. S. & Cheresh, D. A. Integrins in cancer: biological implications

and therapeutic opportunities. Nat Rev Cancer 10, 9-22 (2010).

289 Andrades, J. A. et al. Complement proteins are present in developing

endochondral bone and may mediate cartilage cell death and vascularization.

Exp Cell Res 227, 208-213 (1996).

Page 199: Facultad de Ciencias - CORE

References

170

290 Sakiyama, H. et al. Immunolocalization of complement C1s and matrix

metalloproteinase 9 (92kDa gelatinase/type IV collagenase) in the primary

ossification center of the human femur. Cell Tissue Res 277, 239-245 (1994).

291 Kessenbrock, K., Plaks, V. & Werb, Z. Matrix metalloproteinases: regulators of

the tumor microenvironment. Cell 141, 52-67 (2010).

292 Egeblad, M. & Werb, Z. New functions for the matrix metalloproteinases in

cancer progression. Nat Rev Cancer 2, 161-174 (2002).

293 Ricklin, D. & Lambris, J. D. Complement-targeted therapeutics. Nat Biotechnol

25, 1265-1275 (2007).

294 Rodriguez de Cordoba, S., Harris, C. L., Morgan, B. P. & Llorca, O. Lessons

from functional and structural analyses of disease-associated genetic variants in

the complement alternative pathway. Biochim Biophys Acta 1812, 12-22 (2011).