antileishmanial, cytotoxic and genotoxic effects of

155
ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF ACTINOMYCIN D, Z3, Z5 AND HYDRAZINE DERIVATIVES OF ISOSTEVIOL Ph. D. Thesis By QAISAR JAMAL (M. Sc., M. Phil) DEPARTMENT OF ZOOLOGY UNIVERSITY OF PESHAWAR Session 2013-14

Upload: others

Post on 13-Apr-2022

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

ACTINOMYCIN D, Z3, Z5 AND HYDRAZINE DERIVATIVES OF

ISOSTEVIOL

Ph. D. Thesis

By

QAISAR JAMAL

(M. Sc., M. Phil)

DEPARTMENT OF ZOOLOGY

UNIVERSITY OF PESHAWAR

Session 2013-14

Page 2: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

ACTINOMYCIN D, Z3, Z5 AND HYDRAZINE DERIVATIVES OF

ISOSTEVIOL

Thesis submitted to the University of Peshawar in partial fulfilment of the requirements

for the degree of Doctor of Philosophy in Zoology under the Faculty of Life and

Environmental Sciences by

Qaisar Jamal

DEPARTMENT OF ZOOLOGY

UNIVERSITY OF PESHAWAR

Session 2013-14

Page 3: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

AUTHOR’S DECLARATION

I, Qaisar Jamal Reg. No. 2005-I-2855, student of Doctor of Philosophy in the subject of

Zoology hereby solemnly declare that the subject matter printed in the thesis titled

“Antileishmanial, Cytotoxic and Genotoxic Effects of Actinomycin D, Z3, Z5 And Hydrazine

Derivatives of Isosteviol” is my own work, except wherever stated otherwise and has not been

printed, published and submitted as research work, thesis or publication in any form to any

university or research institution in Pakistan or abroad. If at any stage my declaration is found to

be incorrect even after my graduation, the University of Peshawar will have the right to withdraw

my PhD degree.

Qaisar Jamal

Page 4: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

PLAGIARISM UNDERTAKING

I, Qaisar Jamal Reg. No. 2005-I-2855, student of Doctor of Philosophy in the subject of zoology

hereby solemnly declare that the research work presented in this thesis titled “Antileishmanial,

Cytotoxic and Genotoxic Effects of Actinomycin D, Z3, Z5 and Hydrazine Derivatives of

Isosteviol” is exclusively my own research work, except where stated otherwise. Moreover,

complete thesis has been written by me and wherever any minor contribution/help (if any) was

taken, has been duly acknowledged. I understand the zero-tolerance policy of Higher Education

Commission (HEC), Pakistan and the University of Peshawar towards plagiarism. Therefore, I as

an author of the thesis declare that no portion of my thesis has been plagiarized and any material

used as reference has been properly cited. I undertake that if I am found guilty of any formal

plagiarism in this thesis, the University reserves the right to withdraw/ revoke my PhD degree. I

also declare that HEC and University have the right to publish my name on the HEC/University

website under the list of students with plagiarised thesis.

Qaisar Jamal

Page 5: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

CERTIFICATE OF APPROVAL

This is to certify that the thesis titled “Antileishmanial, Cytotoxic and Genotoxic Effects of

Actinomycin D, Z3, Z5 And Hydrazine Derivatives of Isosteviol” by Qaisar Jamal (Reg. No.

2005-I-2855) under the supervision of Professor Dr. Akram Shah submitted in partial fulfilment

of the requirements for the Doctor of Philosophy (PhD) in Zoology complies with the regulation

of the university to meet the accepted standards with respect to originality.

Student’s Name: Qaisar Jamal _________________________

Examination Committee

External Examiner: 1 _________________________

External Examiner: 2 _________________________

Internal Examiner: 1 _________________________

Supervisor: _________________________

Chairman/Head: _________________________

Page 6: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

RESEARCH COMPLETION CERTIFICATE

It is certified that research work contained in this thesis titled “Antileishmanial, Cytotoxic

and Genotoxic Effects of Actinomycin D, Z3, Z5 And Hydrazine Derivatives of Isosteviol” has

been carried out and completed by Qaisar Jamal (Reg. No. 2005-I-2855) under my supervision. I

recommend it as it fulfills partial requirement for the award of PhD degree in Zoology.

Research Supervisor: _________________________

Professor Dr. Akram Shah

M.Sc. Zoology (Gold Medallist), PhD. (King’s College London)

Commonwealth Post-doctoral Fellowship (School of Medicine, King’s College London)

INSPIRE Postdoctoral Fellow (London School of Hygiene & Tropical Medicine)

Chairman: __________________________

Professor Dr. Akram Shah

Chairman, Department of Zoology

University of Peshawar, 25120 Pakistan

Page 7: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

Dedication

This work is dedicated to Professor Dr. Muhammad Nasim Siddiqui (Late) who worked

sincerely and selflessly to glitter the Science of Zoology in Khyber Pakhtunkhwa Province

of Pakistan. May Allah Bless His Soul in Everlasting Peace and Rehmah, Aameen

Page 8: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

Table of Contents

List of Table………………………………………………………………………………… i

List of Figures……………………………………………………………………………. ii-iv

List of Abbreviations and Symbols……………………………………………………... v-vi

Acknowledgements……………………………………………………………………. vii-viii

Abstract…………………………………………………………………………………... ix-x

1. Introduction………………………………………………………………………... 01

1.1.Cutaneous Leishmaniasis……………………………………………………….. 01

1.2.History and Discovery…………………………………………………………... 02

1.3.Taxonomy and Classification of Leishmania…………………………………… 03

1.4.Evolution of Leishmania………………………………………………………… 07

1.5.Life Cycle and Transmission……………………………………………………. 08

1.6.Epidemiology: Global Scenario………………………………………………… 10

1.7.Surface Molecules Crucial for Survival in Hostile Environments……………… 11

1.8. Genetics of Leishmania………………………………………………………… 12

1.9.Immunology of Leishmaniasis………………………………………………….. 13

1.9.1. Promastigotes in the Inoculum vs Innate Immunity…………………….. 13

1.9.2. Anti-Inflammatory and Immunomodulatory Effect of Sand Fly Saliva… 14

1.9.3. Host Immune Evasion…………………………………………………... 15

1.9.4. The oxidative Burst……………………………………………………... 17

1.9.5. Activation of Natural Killer Cells (NK) and Adaptive Immunity……… 18

1.9.6. Role of Natural Killer Cells…………………………………………….. 18

1.9.7. Th1 and Th2 Response-Mechanism and Outcome……………………... 20

1.10. Treatment Approaches for Leishmaniasis………………………………….. 21

1.10.1. Antimonials…………………………………………………………….. 23

1.10.2. Amphotericin B………………………………………………………… 24

1.10.3. Pentamidine…………………………………………………………….. 26

1.10.4. Miltefosine……………………………………………………………… 27

1.10.5. Paromomycin…………………………………………………………… 28

1.10.6. Phytomedicine in Experimental and Clinical Trials……………………. 28

1.10.7. Inorganic Agents in Experimental and Clinical trials…………………... 33

Page 9: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

1.10.8. Diterpenoids……………………………………………………………. 35

1.10.9. Actinomycins…………………………………………………………... 36

1.11. Starain of Leishmania to be Used………………………………………….. 37

Aim and objectives of the Present Study………………………………………………… 37

2. Morphology and Culture Characterization of Leishmania tropica KWH23…… 38

2.1. Materials and Methods………………………………………………………… 38

2.1.1. Culture of Leishmania tropica KWH23 Promastigotes………………... 38

2.1.2. Evaluation of Morphological Variations of Promastigotes during

Culture………………………………………………………………….. 39

2.1.3. Cultivation of Axenic Amastigotes and Evaluation of Morphology…… 39

2.1.4. Acquisition of Axenic Amastigotes by Starvation……………………... 40

2.1.5. Viability during Transformation………………………………………. 40

2.2. Results…………………………………………………………………………. 41

2.2.1. Growth Curve of L. tropica KWH23 Promastigotes…………………… 41

2.2.2. Axenic Promastigote Morphotypes……………………………………... 42

2.2.3. Division of L. tropica Promastiogtes in Axenic Development…………. 43

2.2.4. Cultivation of Axenic Amastigotes……………………………………... 44

2.2.5. Changes in Outline and Flagella during Differentiation of Promastigotes to

Amastigotes…………………………………………………………….. 45

2.2.6. Viability during Transformation……………………………………….. 49

2.2.7. Natural Acquisition of Axenic Amastigotes……………………………. 50

2.2.8. Requirements for Counting, Fixation and Smearing of Leishmania tropica

KWH23………………………………………………………………….. 51

2.2.9. Revision of Strain Nomenclature………………………………………. 51

2.3. Discussion……………………………………………………………………... 52

3. In Vitro Cytotoxicity Assays of Actinomycins and Isosteviol Derivatives on Human

Peripeheral Blood Lymphocytes

……………………………………………………………………………………… 56

3.1. Materials……………………………………………………………………….. 57

3.1.1. Chemicals and Solutions……………………………………………….. 57

3.1.2. Disposables…………………………………………………………….. 57

Page 10: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

3.1.3. Apparatus………………………………………………………………. 57

3.2. Methods……………………………………………………………………….. 58

3.2.1. Isolation of Human Peripheral Blood Lymphocytes…………………... 58

3.2.2. Inspecting Viability of Lymphocytes………………………………….. 58

3.2.3. Plating Lymphocytes and Adding Compounds……………………….. 59

3.2.4. Evaluation of Cytotoxicity and Cell Death In Vitro…………………… 59

3.2.5. Data Analysis…………………………………………………………... 59

3.3. Results………………………………………………………………………... 60

3.3.1. Cytotoxic Impact of Miltefosine on Lymphocytes……………………. 60

3.3.2. Cytotoxicity of 16 (2,4-Dinitrophenylhydrazine) Isosteviol…………...... 61

3.3.3. Cytotoxicity of 17 Hydroxy, 16 (2,4-dinitrophenylhydrazine)

Isosteviol………………………………………………………………. 62

3.3.4. Cytotoxicity of Benzyl ester, 16 (2,4-dinitrophenylhydrazine)

Isosteviol………………………………………………………………. 62

3.3.5. Cytotoxicity of Actinomycin D………………………………………… 63

3.3.6. Cytotoxicity of Actinomycin Z3………………………………………... 64

3.3.7. Cytotoxicity of Actinomycin Z5………………………………………... 65

3.4.Discussion………………………………………………………………………. 67

4. Antileishmanial Compounds Screening In Vitro……………………………….... 71

4.1.Materials and Methods………………………………………………………….. 72

4.1.1. Culture of the Leishmania tropica KWH23 Promastigotes…………….. 72

4.1.2. Generation of Axenic Amastigotes……………………………………... 72

4.1.3. Antileishmanial Activity………………………………………………... 73

4.1.4. Data Analysis…………………………………………………………… 73

4.2. Results………………………………………………………………………….. 74

4.2.1. Antileishmanial Activity of Miltefosine………………………………... 74

4.2.2. Antileishmanial Activity of 16 (2,4-dinitrophenylhydrazine)

Isosteviol………………………………………………………………... 75

4.2.3. Antileishmanial Activity of 17 hydroxy, 16 (2,4-dinitrophenylhydrazine)

Isosteviol ……………………………………………………………….. 75

Page 11: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

4.2.4. Antileishmanial Activity of Benzyl ester 16 (2,4-dinitrophenylhydrazine)

Isosteviol………………………………………………………………... 76

4.2.5. Antileishmanial Activity of Actinomycin D……………………………. 77

4.2.6. Antileishmanial Activity of Actinomycin Z3………………………….... 78

4.2.7. Antileishmanial Activity of Actinomycin Z5…………………………... 79

4.3.Discussion………………………………………………………………………. 80

4.3.1. Mechanism of Action of Miltefosine…………………………………… 81

4.3.2. Antileishmanial Action of Isosteviol and Actinomycin D……………… 82

5. Evaluation of the Compounds Genotoxicity against Human Lymphocytes……. 86

5.1. Introduction…………………………………………………………………….. 86

5.2. Methodology………………………………………………………………….. 86

5.2.1. Application of Test Compounds to Lymphocytes for Genotoxicity….. 87

5.2.2. Slide Preparation and Mounting of Treated Lymphocytes…………… 87

5.2.3. Lysing of Lymphocyte Cells…………………………………………. 88

5.2.4. Single Cell (SC) Gel Electrophoresis………………………………… 88

5.2.5. Neutralization and Fixation…………………………………………… 88

5.2.6. Staining……………………………………………………………….. 88

5.2.7. Data Interpretation and Scoring………………………………………. 88

5.2.8. Statistical Analysis……………………………………………………. 89

5.3. Results………………………………………………………………………... 90

5.3.1. Genotoxicity of Miltefosine…………………………………………… 90

5.3.2. Genotoxicity of 16 (2, 4-dinitrophenylhydrazine) Isosteviol…………. 91

5.3.3. Genotoxicity of 17 hydroxy, 16 (2, 4-dinitrophenylhydrazine)

Isosteviol………………………………………………………………. 92

5.3.4. Genotoxicity of Benzyl ester 16 (2,4-dinitrophenylhydrazine)

Isosteviol………………………………………………………………. 93

5.3.5. Genotoxicity of Actinomycin D (M1) ………………………………… 94

5.3.6. Genotoxicity of Actinomycin Z3 (M2) ………………………………. 95

5.3.7. Genotoxicity of Actinomycin Z5 (M3) ………………………………. 96

5.4. Discussion……………………………………………………………………. 97

6. General Discussion………………………………………………………………. 99

Page 12: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

Conclusion………………………………………………………………………………... 102

Recommendations………………………………………………………………………... 103

References…………………………………………………………………………….104-133

Page 13: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

i

List of Tables

Table No. Title Page No.

1.1. Classification of Leishmania (modified from Adl et al., 2012; Lukeš et al., 2014) ……….06

1.2. Summary of the genome analysis for five different Leishmania species (Llanes et al., 2015).

……..........................................................................................................................................13

2.1.Promastigote to amastigote transformation ratio over 10-day duration based on Leishmania

morphology.………………………………………………………………………………….47

2.2. Size distribution of various differentiating and transforming amastigotes of Leishmania

tropica KWH23 in acidified medium……………………………………………………........48

2.3. Size distribution of the completely round, intermediates and oval forms of Leishmania

tropica amastigotes.………………………………………………………….........................48

Page 14: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

ii

List of Figures

Figure No. Title Page No.

Figure 1.1. Classification of Leishmania (Adl et al., 2012; Lukeš et al., 2014) ……………05

Figure 1.2. Geographical distributions of various Leishmania spp.; sandflies and animal

reservoirs in the Old and New World. L: Leishmania (species), S: Sandfly (genus

or subgenus), R: Reservoir (genus or family) (Akhoundi et al., 2016)………….08

Figure 1.3. Life cycle of Leishmania showing different developmental stages of promastigote

in the sand fly midgut. The figures are from Ortiago et al., 2010 (A), Oliveira et

al., 2009 (B) and Dostálová and Volf, 2012 (C). ……………………………….10

Figure 1.4. Difference between the LPG of procyclics and metacyclics (A) and species-

specific variability in structure of LPG structure of other major glycoconjugates

(B). (A) from Spath et al., (2001) and (B) from Kamhavi, (2006) …………….12

Figure 1.5. Immunomodulation mechanism in macrophages (Bogdan and Röllinghoff, 1998)

…………………………………………………………………………………...17

Figure 1.6. Activation of natural killer cells in experimental visceral leishmaniasis (Bogdan et

al., 2012) ………………………………………………………………………...19

Figure 1.7. Immune response in cutaneous Leishmania infection (Abdossamadi et al., 2016)

…………………………………………………………………………………..20

Figure 2.1. Hemocytometer illustration representing set of 16 squares (marked in red colour)

used for counting promastigotes………………………………………………..38

Figure 2.2. Representative growth curve of Leishmania tropica KWH23 in vitro………… 41

Figure 2.3. Different stages of L. tropica KWH23 promastigotes representing cell cycle in

culture (see text for description). A naturally occurring amastigote (Arrow in “e”)

can be seen in a 13-day old promastigote culture………………………………42

Figure 2.4. Division of the L. tropica promastigotes in axenic development. Cells with 2

flagella single kinetoplast and single nucleus (A and B), Cells with single

flagellum, two kinetoplasts and single nucleus (C), Cells with Single flagellum,

two kinetoplasts and two nuclei (D and E) and Cells with two flagella, two

kinetoplasts and two nuclei near to the completion of cytokinesis (F). Solid

arrows show kinetoplasts and the others indicate flagella………………………44

Page 15: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

iii

Figure 2.5. Promastigote to amastigote transformation. Change in the cell morphology and

loss of flagellum is seen progressively…………………………………………45

Figure 2.6. Viability assessment for Leishmania tropica amastigotes during transformation

and differentiation………………………………………………………………49

Figure 2.7. Unsurprisingly acquired amastigotes (A), Starved culture medium with pH 4.8-5.0

having naturally changed amastigotes (B). Standard RPMI 1640 growth medium

with pink colour and phenol red as a pH indicator (C)………………………….50

Figure 3.1. Concentration dependent cytotoxicity of Miltefosine against human peripheral

blood lymphocytes……………………………………………………………….61

Figure 3.2. Cytotoxicity of 16 (2,4-dinitrophenylhydrazine) Isosteviol against human

peripheral blood lymphocytes …………………………………………………...62

Figure 3.3. Cytotoxicity of 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol against

human peripheral blood lymphocytes……………………………………………63

Figure 3.4. Cytotoxicity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol against

human peripheral blood lymphocytes……………………………………………64

Figure 3.5. Cytotoxicity of Actinomycin D against human peripheral blood lymphocytes…65

Figure 3.6. Cytotoxicity of Actinomycin Z3 against human peripheral blood lymphocytes...66

Figure 3.7. Cytotoxicity of Actinomycin Z5 against human peripheral blood lymphocytes...67

Figure 4.1. Cutaneous leishmaniasis patients are treated in Peshawar Hospital with

intralesional injection of meglumine antimonate………………………………..71

Figure 4.2. Antileishmanial activity of miltefosine against promastigotes (A) and amastigotes

(B) of L. tropica KWH23……………………………………………………….74

Figure 4.3. Leishmanicidal activity of 16 (2,4-dinitrophenylhydrazine) Isosteviol against

promastigotes (A) and amastigotes (B) of L. tropica KWH23………………….75

Figure 4.4. Antileishmanial activity of 17 hydroxy, 16 (2,4-dinitrophenylhydrazine)

Isosteviol against promastigotes (A) and amastigotes (B) of L. tropica KWH23.76

Figure 4.5. Antileishmanial Activity of Benzyl ester 16 (2,4-dinitrophenylhydrazine)

Isosteviol against promastigotes (A) and amastigotes (B) of L. tropica KWH23.77

Figure 4.6. Antileishmanial activity of Actinomycin D (M1) against promastigotes (A) and

amastigotes (B) of L. tropica KWH23…………………………………………..78

Page 16: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

iv

Figure 4.7. Antileishmanial activity of Actinomycin Z3 (M2) against promastigotes (A) and

amastigotes (B) of L. tropica KWH23…………………………………………..79

Figure 4.8. Antileishmanial activity of Actinomycin Z5 (M3) against the promastigotes (A)

and amastigotes (B) of L. tropica KWH23………………………………………80

Figure 4.9. Leishmania promastigotes (A) and amastigotes (B) showing acidocalcisomes

(https://www.sciencedirect.com/science/article/pii/S0020751907001038#fig1)..82

Figure 5.1. Genotoxicity of Miltefosine (Milt) at 100 uM, 25 uM and 1.25 uM (IC50 for

Leishmania tropica)…………………………………………………………….90

Figure 5.2. Genotoxicity of 16 (2,4-dinitrophenylhydrazine) isosteviol (ID) at 100 uM, 25 uM

and 4.447 uM (IC50 for Leishmania tropica)…………………………………...91

Figure 5.3. Genotoxicity of (ID7) at 100 uM, 25 uM and 5.033 uM (IC50 for Leishmania

tropica)………………………………………………………………………….92

Figure 5.4. Genotoxicity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) isosteviol (IDB5) at

100 uM, 25 uM and (IC50 for Leishmania tropica)…………………………….94

Figure 5.5. Genotoxicity of Actinomycin D (M1) at 100 uM, 25 uM and 2.135 uM (IC50 for

Leishmania tropica)……………………………………………………………..95

Figure 5.6. Genotoxicity of Actinomycin Z3 (M2) at 100 uM, 25 uM and 1.76 μM IC50 for

Leishmania tropica……………………………………………………………...96

Figure 5.7. Genotoxicity of Actinomycin Z5 (M3) at 100 uM, 25 uM and 1.691 μM (IC50 for

Leishmania tropica)……………………………………………………………..97

Page 17: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

v

List of Abbreviations and Symbols

ANOVA Analysis of variance

CR Complement receptor

DMSO Dimethyl Sulfoxide

EBVF Elongated barely visible flagellum

EDTA Ethylene diamine tetra-acetic acid

ELF Elongated long flagellum,

ENF Elongated no flagellum

ESF Elongated short flagellum

GP63 Metalloproteinase glycoprotein 63

GIPL Glycoionositolphospholipid

hi-FBS Heat-inactivated fetal bovine serum

HI-FCS Heat-inactivated fetal calf serum

ID 16 (2,4-Dinitrophenylhydrazine) Isosteviol

ID7 17 Hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol

IDB5 Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol

IFN-γ Interferon Gamma

IL-10 Interleukin-10

IMS Industrial methylated spirit

IU International Unit

KWH23 Kuwait Teaching Hospital (Original ID of the strain of Leishmania

tropica)

LPG Liphophosphoglycan

M1 Actinomycin D

M2 Actinomycin Z3

M3 Actinomycin Z5

MIC Minimal inhibitory concentration

Page 18: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

vi

Milt Miltefosine

mL Millilitre

Mɸ Macrophage

OBVF Ovoid barely visible flagellum

OLF Ovoid long flagellum

ONF Ovoid no flagellum

OSF Ovoid short flagellum

PBLs Peripheral blood lymphocytes

PBS Phosphate Buffer Saline

PCD Programmed cell death

Plain Negative control (Cells of parasites grown in medium only)

RBVF Rounded barely visible flagellum

RLF Rounded long flagellum

RNF Rounded no flagellum

RPMI-1640 Rosewell Park Memorial Institute (culture medium)

RSF Rounded short flagellum

SSG Sodium Stibogluconate

TGF-β Transforming Growth Factor-Beta

XTT 2,3-Bis-(2-Methoxy-4-Nitro-5-Sulfophenyl)-2H-Tetrazolium-5-

Carboxanilide

μM Micromolar

μm Micrometer

Page 19: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

vii

Acknowledgements

I am greatly indebted to my research supervisor Professor Dr. Akram Shah whose support

accompanied me all the way long during my M.Sc., M.Phil and now Ph.D. Had he not been that

much generous to me, perhaps I won’t have made it possible to go through the worries of

research work. His sincere efforts and dedication did not let me, and many others, feel any dearth

of materials and equipment. All this was to make things easier for me and point me in the course

of the light at the end of the channel.

I am thankful to Dr. Syed Basit Rasheed, Assistant Professor Department of Zoology, University

of Peshawar for his wise and insightful suggestions during experimental work. His wholehearted

response was extremely encouraging and helpful during jammed moments of laboratory work.

He, being much busy investigator, never said a nope, when requested for assistance. His

guidance in writing the manuscript is worth salutation.

I will not forget enthusiastic help from Dr. Shumaila Noreen, Assistant Professor Department of

Zoology, University of Peshawar when she lent me the sum to pay my fees. Rather, it was this

onward that I could complete my official documentation necessary in academic degrees. Thanks

to her for being generous enough to cut out money from her expenditures during this growing

inflation to help me.

I really owe to acknowledge my hostel mate Mr. Ibadat Ur Rehman, then Lecturer Hakim Abdul

Jalil Nadvi University College for Boys, University of Peshawar and now Junior Scientist

Pakistan Atomic Energy Commission, for suggesting me to use ortho-phosphoric acid for the

acidification of medium for Leishmania amastigotes culture; that worked wonderfully.

My heartfelt thanks go to Dr. Sanaullah Khan, Associate Professor Department of Zoology,

University of Peshawar for his concern and provoking advices, on occasions, to pursue my Ph.D

work on priority.

I am also much grateful to Dr. Asad Ullah, Assistant Professor Department of Chemistry, Islamia

College Peshawar for providing me the test compounds. He has always been generous and

gracious enough to provide guidance and information on the compounds. His ready response will

always be remembered.

Page 20: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

viii

I feel pleasure to express my gratitude to Dr. Muhammad Khisroon, Assistant Professor

Department od Zoology, University of Peshawar for his support during genotoxicity evaluation.

His efforts in establishing and optimizing the comet assay techniques in the Department of

Zoology, University of Peshawar are clear like broad day light. He has been generous enough in

providing material, technical and theoretical support during these assays. He also went through

that part of the manuscript and put in valuable suggestions.

Valuable suggestions and faithful advices from Dr. Sobia Wahid and Dr. Nazma Habib,

Assistant Professors Department of Zoology, are worth acknowledgment. We were lucky to use

Leishmania tropica KWH23 (MHOM/PK/2010/KWH23) collected and typed by Dr. Nazma

Habib during her PhD at London School of Hygiene & Tropical Medicine. Madam thanks for

opening this door of opportunity.

Thanks are also due to Mr. Zaigham Hasan and Dr. Farrah Zaidi, Dr. Mohammad Adnan,

Assistant Professors, Department of Zoology for advices, support and assistance.

I heartily acknowledge ministerial help from Mr. Farhat Ullah, Senior Assistant, Department of

Zoology, University of Peshawar and Ashfaq Ali, Junior Clerk Department of Zoology,

University of Peshawar.

Last, but not least, I feel privileged to thank my fellow PhD scholars Mr Ajmal Khan and Mr

Abid Ullah who helped me selflessly in running comet assay protocol. They provided me basic

understanding and practical handling of the technique.

Qaisar Jamal

Page 21: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

ix

Abstract

In the present study in vitro culture of Leishmania tropica KWH23 (MHOM/PK/2010/KWH23),

was used for all the experiments. In axenic growth, Leishmania tropica promastigotes reached to

log, mid-log, late-log and stationary phases on day 4, 5, 6 and 7 in culture respectively. Among

stationary phase promastigotes higher density of metacyclic was reported. Nectomonad stage

promastigotes were found to be the longest and slender most individuals outnumbering any other

morphotype for the first three days in the culture. On day 3 onward, leptomonads appeared in the

culture to a ratio of 44%. They were distinguished from nectomondas by getting wider anteriorly.

Metacyclic promastigotes appeared in culture during log phase with metacyclic to leptomonad to

nectomonad ratio of 27, 43 and 29% respectively. During logarithmic growth, 17% of the

promastigotes were found to be dividing. Division normally proceeded from flagellum to

kinetoplast to nucleus. Amastigote stage was grown in vitro in axenic culture. Day 4 onward

most of the parasites in culture were represented by rounded and ovoid cells with no flagella.

Cell size decreased from 10.966μm of the promastigote to 3.138μm of round amastigote. During

the transformation process 96-98% viability was noted. When the promastigotes were left

without fresh medium change, they naturally changed to amastigotes due to pH drop. In a 10 day

follow up, the pH dropped from 7.4 to 4.8 and 91% of the parasites, at a density of 1.2x107,

changed to amastigotes having 97% viability. These amastigotes were successfully transformed

back to promastigotes in normal growth medium.

Antileishmanial, cytotoxic and genotoxic effects of Actinomycin D, Z3 and Z5 and Isosteviol

derivatives, 16 (2,4-dinitrophenylhydrazine) Isosteviol, 17-hydroxy 16 (2,4-

dinitrophenylhydrazine) Isosteviol and Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol

were assessed. Miltefosine was used as standard positive control. Cytotoxicity was expressed i]n

terms of 50% inhibitory (IC50) values. The IC50 values of Miltefosine, 16 (2,4-

dinitrophenylhydrazine) Isosteviol, 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol,

Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol, actinomycin D, actinomycin Z3 and

actinomycin Z5 were 272.2μM (95 % CI= 143.6 to 515.7μM), 781.2μM (95 % CI= 240.8 to

2535.0μM), 294.1μM (95 % CI= 177.4 to 487.5μM), 421.8μM (95% CI= 211.3 to 842.1μM),

195.8μM (95% CI=135.3 to 283.2μM), 210.1μM (95% CI= 145.2 to 304.1μM), and 234.9μM

(95% CI= 155.5 to 354.9μM) respectively regarding cytotoxicity.

Page 22: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

x

Regarding antileishmanial activity, Miltefosine, 16 (2,4-dinitrophenylhydrazine) Isosteviol, 17-

hydroxy 16 (2,4-dinitrophenylhydrazine) Isosteviol, Benzyl ester 16 (2,4-

dinitrophenylhydrazine) Isosteviol, Actinomycin D, Actinomycin Z3 and Actinomycin Z5 gave

IC50 values against L. tropica promastigotes and amastigotes as 10.80μM (95% CI=9.114 to

12.81μM), 1.245μM (95% CI=0.7250 to 2.138μM), 7.098μM (95% CI=5.328 to 9.455μM),

4.447μM (95% CI= 2.788 to 7.094μM), 5.603μM (95% CI= 4.628 to 6.784μM), 5.033μM (95%

CI= 3.189 to 7.945μM), 10.71μM (95% CI= 8.611 to 13.31), 6.794μM (95% CI= 4.248 to

10.87μM), 8.739μM (95% CI= 6.675 to 11.44μM), 2.135μM (95% CI= 1.419 to 3.211μM),

5.500μM (95% CI= 3.811 to 7.939μM), 1.760μM (1.136 to 2.728μM), 9.529μM (95% CI= 7.354

to 12.35μM) and 1.691μM (95% CI= 0.9559 to 2.991μM) respectively. In conclusion, L. tropica

KWH23 was extra sensitive to 16 (2,4-dinitrophenylhydrazine) Isosteviol.

Miltefosine gave least genotoxicity at 100, 25 and 1.25μM concentration having total comet

score (TCS) of 10, 8 and 8 respectively. Damage was non-significant (P>0.05) as compared to

1% DMSO and negative control. Compound 16 (2,4-dinitrophenylhydrazine) Isosteviol showed

concentration dependent genotoxicity. It gave TCS values of 207.33, 87.33 and 10.66

respectively at 100, 25 and 4.447μM concentration. The compound showed non-significant

genotoxic effects to the standard Miltefosine and 1% DMSO and negative control at all the

concentration (P>0.05). Compound 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol

caused significant genotoxicity as compared to standard and negative control at 100 and 25μM

(P<0.05). At 5.033μM concentraton, however, the genotoxicity became non-significant (P>0.05).

Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol was found to be non-genotoxic as

contrasted with the standard and negative control at all concentrations (P>0.05). The TCS values

calculated were 166.33, 85.33 and 15 respectively at 100, 25 and 6.794μM concentrations.

Actinomycin D showed highest degree of genotoxicity as compared to the standard, Isosteviol

derivatives and negative control at 100 and 25μM concentrations (P<0.05). But Genotoxicity

became non-significant at 2.135μM concentrations (P>0.05). Actinomycin Z3 was found to show

significant genotoxicity at all the concentration i.e., 100, 25 and 1.76μM in relation to standard

and negative control as well as Isosteviol derivatives (P<0.05). Actinomycin Z5 was also found

significantly genotoxic as compared to standard, negative control and Isosteviol derivatives at all

the concentration (P<0.05). In terms of TCS values the genotoxicity, however, greatly reduced

with decreasing concentration 100μM (337.666), 25μM (214.333), 1.691μM (23.666).

Page 23: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

1

CHAPTER 1. INTRODUCTION

Leishmaniasis is a collective term for vector-borne diseases (Saunkratay et al., 2010) affecting

mainly the skin (cutaneous leishmaniasis), mucous membranes (muco-cutaneous leishmaniasis)

and reticulo-endothelial system (visceral leishmaniasis) of vertebrates including human (WHO,

2019). Obligate intracellular protozoans in the genus Leishmania (Order: Kinetoplatida; Family:

Trypanosomatidae) are the causative agents of the diseases (Gupta and Nishi, 2010; Hatam et al.,

2013; Llanes et al., 2015). Clinical manifestations of the infection show variability in relation to

the causative species and site of infection. Even same species may reflect different clinical

history in relation to different individuals, as well as hosts and geography (Auon and Bouratbine,

2014). Due to the fact, the categorization of Leishmania species to visceral and cutaneous in

humans is infrequently applicable in other mammalian hosts e.g., Leishmania infantum infection

in dogs has been found to be viscero-dermic and human cutaneous Leishmania species have been

recovered from rodent’s viscera (Roque and Jansen, 2014). Leishmaniasis is principally zoonotic

except Leishmania donovani and Leishmania tropica, though few reports suggest animal

reservoirs for these two Leishmania species across Asia and Africa (Burza et al., 2018).

1.1. Cutaneous Leishmaniasis

WHO estimates 0·7–1 million cutaneous leishmaniasis (CL) cases yearly in the world. Presently,

more than 90% of CL cases are presented in Afghanistan, Algeria, Brazil, Iran, Pakistan, Peru

Saudi Arabia and Syria. Due to cross-border migration of war refugees, the number of CL cases

has noticeably amplified in non-endemic republics. Leishmania species producing CL in Old

World are Leishmania major, L. tropica, and L. aethiopica, that are predominant round the Horn

of Africa, Middle East, Mediterranean Basin and the Indian Subcontinent. The New World CL

causing Leishmania species includes Leishmania amazonensis, L. braziliensis, L. guyanensis and

L. mexicana reported from Central and South America (Burza et al., 2018).

Cutaneous leishmaniasis shows variety in clinical presentation of signs and symptoms vacillating

from simple to diffuse forms. Simple form of the disease is characterized by single or multiple

localized, self- healing and painless skin lesions developed at the site of sandfly bite. In worse

cases, it can cause disfiguring scars leading to social stigmatization. Diffuse form demonstrates

nodular lesions dispersed over the body (Debrant et al., 2004; Bahrami et al., 2011; Corrêa et al.,

Page 24: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

2

2005). With reference to CL distribution sandfly vector belonging to Genus Phlebotomus is

found in the Old World and Lutzomyia in the New World. Vectors other than phlebotomine

sandflies have recently been incriminated. Day feeding hematophagous midges in subgenus

Forcepomyia (Lasiohelea) Keifer have been found to transmit a Leishmania species (not

properly identified but closest to L. enrietii) in red kangaroo (Macropus rufus) in Australia

(Dougall et al., 2011). CL is either anthroponotic (L tropica) with human-to-human infection or

zoonotic (L aethiopica, L major and all species found in the New World) that can be transmitted

from animals to human (Burza et al., 2018; Soptin et al., 2015). Rodents (Order: Rodentia),

carnivores (Order: Carnivora), marsupials such as opossums (Order: Didelphimorphia), anteaters

and sloths (Order: Pilosa), armadillos (Order: Cingulata) and primates (Order: Primata) have

been incriminated major reservoirs in zoonotic CL reported in various parts of the world (Bari

and Rahman, 2008; Roque and Jansen, 2014).

1.2. History and Discovery

Leishmaniasis is an ancient disease which is presented as historical figures, statues, ceramics and

papyrus (Tuon et al., 2008). The presence of Leishmania-like species in prehistorical eras is

predicted in two fossil ambers. The first Leishmania-like fossil was reported from the proboscis

and alimentary region of a blood-packed vanished female sand fly Palaeomyia burmitis

preserved in a 100 million-year old Cretaceous Burmese amber. The second fossil was described

as Paleoleishmania neotropicum originated in the extinct sand fly Lutzomyia adiketis in a 20–30

million-year-old Dominican amber. Leishmania promastigotes and amastigotes were perceived

in the gut and proboscis of the sand fly; though, no human or animals blood cells were

recognized. Leishmania species has perhaps evolved in Mesozoic era (252–66 million years ago)

earlier to the disintegration of the supercontinent Pangaea. Though, the precise geographical

establishment of the Leishmania species is a substance of continuing discussion (Steverding,

2017).

Ancient history of leishmaniasis has been traced back from 1500-2500 BC based on explanation

of Old-World CL lesions on medications in the library of King Ashurbanipal from 7th century

BC and DNA studies from Egyptian mummies and other archaeological remains. Recent records

are, however, found with 10th century Persian Muslim physician, astronomer, thinker and writer

of the Islamic Golden Age Avicenna (Abu 'Ali al-Husayn ibni Sina) who gave a detailed

Page 25: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

3

description of the then and now called Balkh sore from Northern Afghanistan. Later records of

cutaneous leishmaniasis from Middle East were given mostly with local names of the disease

referring to the cutaneous lesions. Ancient history of visceral leishmaniasis or “kala azar” is not

clearly recognized. It was first noticed in District Jessore in India in 1824 but was mistaken for a

virulent form of malaria even by Ronald Ross. Original explanation of kala-azar was by the army

doctor William Twining (1790–1835) when he published a paper in 1827 regarding patients in

Bengal, India, who seemed withered with splenomegaly, severe anemia and sporadic

temperature. The initial epidemic of kala-azar was documented in 1824-25 in village

Muhammedpore thirty miles east of Jessore with mortality rate of 30%. Its status as a different

disease remained hidden until the discovery of the causative agent of visceral form of the disease

(Leishmania donovani) independently by Scottish army doctor, William Leishman and a

Professor of physiology at Madras University, Charles Donovan in 1900 (Steverding, 2017).

Scottish doctor and naturalist Alexander Russell (1715–1768) in 1756 published a

comprehensive clinical explanation of both dry and wet forms of Oriental lesion when he was

working in Aleppo city of Syria. He assigned CL as wet zoonotic CL instigated by L. major and

dry anthroponotic CL produced by L. tropica with mercurial plaster as successful treatment for

both types (Steverding, 2017). A number of people such as American James Homer Wright,

however, had seen parasites of cutaneous form of the disease but could not have properly

described. It was in 1885 and 1898 when David Cunningham and a Russian military doctor,

Borosky respectively described the Leishmania parasite as what really it is (Cox, 2002).

1.3. Taxonomy and Classification of Leishmania

An accurate classification and systematic account that depicts a clear picture of the phylogeny

and evolutionary history is normally crucial in elucidating its biology, relationship with host and

identification of strains. Morphology has been the main aspect of classification of almost all

groups of parasites until very recent years. However, trustworthiness of morphological

characterization is mostly open to errors because of plasticity of morphological characteristics.

With the beginning of molecular techniques, the fields of systematics and phylogenetics have got

into new direction. Molecular approaches are now excessively used to revise the classifications

done using traditional methods. Molecular systematics is unraveling many curiosities regarding

Page 26: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

4

host-parasite interactions by exploring various aspects of the biology of serious human and

animal parasites (Monis, 1999).

Although, after its first description in 1903, much work has been done regarding genus

Leishmania, however, the evolutionary history and taxonomic classification of its species

remains incomplete and debatable. Confusion in the hierarchic classification of the taxon exists

at the level of genus and lower categories. This conflicting situation and uncertainty regarding

the taxonomy of the taxon is mainly due to incompatibility of molecular and non-molecular data

(based on clinical manifestations, biology of the parasites, geographic distribution, epidemiology

and immunological and biochemical characteristics) as well as studies being inclined to mostly

human infecting species (Harkins et al., 2016; Schönian et al., 2010).

Currently, the 35 to 40 (Fraga et al., 2013; Lukeš et al., 2014) different species in genus

Leishmania (Family: Trypanosomatidae: Order: Kinetoplastea) have been divided into two

sections Euleishmania and Paraleishmania (Ramalho-Ortiago et al., 2010). The section

Euleishmania contains species in three sub-genera, Leishmania, Viannia and Sauroleishmania

(Momen and Cupolillo, 2000; Fraga et al., 2010) and a separate new subgenus described in

Ghana L. enrietti complex. Species infecting humans belong to the former two sub-genera and L.

enrietti complex. The division of human Leishmania species into two sub genera originally was

done by Lainson and Shaw (1987) and was based on the region-specific growth and development

in the gut of sand fly vector. Members of the former prefer to develop in the midgut and pyloric

region (suprapylarian development) while that of the latter in the hindgut (peripylarian

development). This division is now supported by molecular data especially study of Fraga et al.

(2010) using molecular marker heat-shock protein 70 gene. L enrietti complex consists of four

species L. enrietti itself, L. siamensis, L. martiquensis and an unnamed species from Australia

(Llanes et al., 2015; Kwakye-Nuako et al., 2015; Corrêa et al., 2005). Species that infect Old

World reptiles and have been isolated from several lizards are classified in a separate subgenus

of Leishmania called as Sauroleishmania. Status of Sauroleishmania has, however, remained

conflicting among the workers. Some of the workers consider it as a separate genus and do not

place it in either of the sections (Momen and Cupolillo, 2000; Cupolillo et al., 2000).

Sauroleishmania (in whatever as a separate genus or as a subgenus) comprises of four different

species L. (S.) hoogstraali, L. (S.) adleri, L. (S.) gymnodactyli and L. (S.) tarantolae (Kwakye-

Page 27: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

5

Nuako et al., 2015). Paraleishmania includes Leishmania hertigi and L. deanei that infect

porcupines and L. colombensis, L. equatorensis and L. herreri of sloths and squirrels and an

apparently unclear group, usually considered a genus, Endotrypanum. Endotrypanum currently

has two species E. shaudini and E. monterogeii. L. colombensis has been isolated from many

different hosts including humans (Momen and Cupolillo, 2000; Cupolillo et al., 2000).

Figure 1.1. Classification of Leishmania (Adl et al., 2012; Lukeš et al., 2014).

Page 28: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

6

Table 1.1. Classification of Leishmania (modified from Adl et al., 2012; Lukeš et al., 2014)

Section Complex Species Distribution

Euleishmania Leishmania donovani (OW) L. (L.) donovani OW

L. (L.) archibaldi OW

1. L. (L.) infantum 2. Syn. L. (L.) chagasi 1= OW, 2= NW

Leishmania tropica (OW) L. (L.) tropica OW

L. (L.) aethiopica OW

L. (L.) killicki OW

Leishmania major (OW) L. (L.) major OW

L. (L.) gerbilli OW

L. (L.) arabica OW

L. (L.) turanica OW

Leishmania amazonensis (NW) L. (L.) mexicana (syn. L. (L.) pifanoi) NW

L. (L.) amazonensis (Syn. L. (L.)

gamhami ) NW

L. (L.) aristidesi NW

L. (L.) venezuelensis NW

L. (L.) forattinii NW

Leishmania enrietti (OW) L. enrietti OW

L. siamensis OW

L. martiniquensis OW

Leishmania brasiliensis (NW) L. (V.) brasiliensis NW

L. (V.) peruviana NW

Leishmania guyanensis (NW) L. (V.) guyanensis NW

L. (V.) panamensis NW

L. (V.) shawi NW

L. (Vianna) sp not recognized to

complex L. (V.) naiffi NW

L. (V.) lainsoni NW

L. (V.) lindenbergi NW

L. (V.) utingensis NW

L. (S.) hoogstraali OW

(L. S.) adleri OW

L. (S.) gymnodactyli OW

L. (S.) tarantolae OW

Paraleishmania

L. colombiensis NW

L. equatorensis NW

L. herreri NW

L. hertigi NW

L. deanei NW

Page 29: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

7

1.4. Evolution of Leishmania

The phylogeny of genus Leishmania has been established on both the traditional (vector

specificity, clinical picture, geographic distribution, reservoir) as well as molecular data but it

still remains incomplete and unanswered (Marcili et al., 2014). Based on eco-biological aspects

Leishmania evolution has been influnced by its reservoirs and vector hosts. Its has been

speculated to be evolving from an estimated one billion years (der Auwera et al., 2011). Fossil

record indicates that Excavata, a group to which euglinids and kinetoplastids belong, have

eveolved in the Ordovician epoch of Palaeozoic era. The primitive most ancestor of the current

Leishmania might not have evolved until 700 million years ago in Proterozoic eon, where the

kingdom animalia (metazoans) could have evolved to provide host (both vector and deinitive) for

the parasites. The primitive host could have been a water dwelling animal. The Ordovician

ancestors of the Leishmania might have been parasitizing fish and amphibians via leeches. The

evolution of winged insects may be 300 miilion years ago and appearance of primitive winged

insets in diptera (current vectors of Leishmania) might have facillitated divergence from

primitive to new forms of Leishmania. Recovery of fossil haematophagus winged insects from

cretaceous about 140 miilion years ago and ancestral Palaeoleishmania proteus from again

cretaceous some 100 miilion years ago reveals the ingestion of trypanosomatid like free living

flagellates by cretaceous sandfly larvae, propagating them to adults and transmiting to the

vertebrates; initiating the life cycle. The descendants for current Leishmania species might have

evolved in paleogene period about 50 million years ago (Tuon et al., 2008).

Several hypotheses have been proposed for the origin of Leishmania regarding geographic

regions but the conflict still exists. Based on the previously mentioned non-molecular data,

Leishmania is hypothesized to have evolved in palaearctic lizards with Sauroleishmania being a

sister clade. According to this criteria a subsequent spread of Leishmania to Nearctic and

Neotroic through land bridges is assumed. Recent molecular approaches, however, support a

Neotropical origin of the parasite and subsequent spread to the Palearctic and Nearctic reigions

of the world (Tuon et al., 2008).

Page 30: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

8

Figure 1.2. Geographical distributions of various Leishmania spp.; sandflies and animal

reservoirs in the Old World and New World. L: Leishmania (species), S: Sandfly (genus or

subgenus), R: Reservoir (genus or family) (Akhoundi et al., 2016).

1.5. Life Cycle and Transmission

All species of Leishmania possess a digenetic life cycle in which they move between an

invertebrate vector (sand fly) and definitive vertebrate host. Females of about 30 different

species (Lukeš et al., 2007) of hematophagous sand flies belonging to Genus Phlebotomus in the

Old World and Lutzomyia in the New World, are primary vectors of all kinds of human

leishmaniasis around the world. Species in the subgenus Sauroleishmania are transmitted to

lizard by sand flies in genus Sergentomyia (Momen and Cupollilo, 2000). The life cycle begins

when a female fly takes blood meal from an infected mammalian or other vertebrate host. The

fly ingests infected macrophages containing the non-flagellated form of the parasite called

amastigote in their phagolysosomes. When the macrophages lyse in the gut of the sand fly, the

amastigote form emerges and is soon transformed to a flagellated or promastigote form. The

Page 31: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

9

transformation from amastigote to promastigote takes about 4 to 7 days or sometimes 10 days

depending on species and is accompanied by several intermediate stages of the promastigote to

the final infective metacyclic form. Stimuli that triggers the amastigote to promastigote form

have been reported to be lower temperature (from 37oC to 24oC) and high pH (from 7.3 to 8.5) of

the sandfly gut. During early days after blood suckling, natural obstacles to Leishmania growth

comprise proteolytic enzymes, the peritrophic medium adjacent the swallowed blood meal and

sandfly’s immune response. During the transformation cycle, the parasite has to face several

challenges and barriers to reach the final stage. Having an appropriate adaptation to travel a safe

journey through this critical intermediary stage of transformation is necessary for the parasite to

avoid being perished otherwise. The early promastigotes, the weakly motile long procyclic stage

with short flagellum, remain for 2-3 days in the blood meal enclosed by the peritrophic matrix

(PM) secreted by the mucosa of the sand fly’s gut. At the emergence on the decomposition of

PM, the very first priority of the strongly motile long promastigote, now called nectomonads,

move toward the anterior midgut (in case of subgenus Leishmania (Leishmania) which is called

suprapylarian with reference to this anterior movement as against the peripylarian subgenus

Leishmania (Viannia) that enters the hindgut after emergence from PM and then come to

midgut). The nectomonad stage transform to short leptomonad stage (Figure 1.4) now in the

anterior midgut (Gossage et al., 2003; Oliveira et al., 2009; Ramalho-Ortiago et al., 2010).

Page 32: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

10

Figure 1.3. Life cycle of Leishmania species showing different developmental stages* of

promastigote in the sandfly midgut (adopted from Ramalho-Ortiago et al., 2010 (A), Oliveira et

al., 2009 (B) and Dostálová and Volf, 2012 (C).

*The figures display amastigote to procyclic and nectomonad development which occurs in the blood meal enclosed

in peritrophic matrix (PM). Nondividing nectomonad emergence from PM is followed after its attachment to the

epithelium of the gut and its change to leptomonad stage. The leptomonad develops to haptomonads and then to the

infectious metacyclic stage.

1.6. Epidemiology: Global Scenario

Ninety-nine countries and three territories of the world in five different continents (Africa, South

America, Europe, Asia and Australia) embrace leishmaniasis (Fraga et al., 2013; Strekers et al.,

2014; Rodrigues et al., 2015). An estimated 350 million people are at risk of the disease and 12

million are currently affected. The annual incidence of the disease is estimated to be

approximately 2 million cases worldwide (Carriόn et al., 2011) with a break up of 0.2- 0.4

million and 0.7-1.2 million cases of visceral and cutaneous leishmaniasis respectively (Akhoundi

Page 33: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

11

et al., 2016). The actual reported cases are however, 0.6 million (Rodrigues et al., 2015). Annual

death rate, due to visceral infection is 30,000-50,000 cases worldwide (WHO, 2018). Although

the disease is widely distributed around the world, majority of the disease incidence occurs in a

limited number of countries. Almost 90 % of the cases of visceral leishmaniasis are reported

from only six countries that include India, Sudan, South Sudan, Ethiopia, Bangladesh and Brazil.

For cutaneous leishmaniasis, 70-75 % of the cases come from ten countries including

Afghanistan, Algeria, Colombia, Ethiopia, Brazil, Iran, Syria, North Sudan, Costa Rica and Peru.

In most cases, leishmaniases are zoonoses mostly affecting poor people in the rural areas.

Domestic dogs have been proved as potential reservoir for human leishmaniasis and 13 of 21

human Leishmania species have been so far isolated from dogs (Beattie and Kaye, 2011;

Cantasessi et al., 2015; WHO, 2018).

1.7. Surface Molecules Crucial for Survival in Hostile Environments

Some of the surface molecules play important role in the parasite’s overall biology. Important

most of these are glycoconjugates phosphoglycans including liphophosphoglycan (LPG),

metalloproteinase glycoprotein 63 (gp63) also called leishmanolysin, secreted acid phosphatase

(SAP) and filamentous (fPPG) proteophosphoglycan (Kamhavi, 2006). All these are accounted

for adaptability of the Leishmania to avert and survive the adversities presented by the hostile

environments of both the vector and definitive host and are important virulence factors. LPG,

however, is at the peak of prestige in fighting against the hostilities of the hosts. This molecule is

involved in avoidance of being digested by sandfly gut enzymes, scavenging of free radicals of

oxygen in the blood meal, prevention of procyclic and nectomonad stages from being flushed out

with digested blood remnants (Vanier-Santos et al., 2002). Along with gp63, internalization of

metacyclics by macrophage occurs through complement receptors CR1, CR3, mannose receptors

(MR) and fibronectin (FnR) receptors (Teixeira et al., 2013).

Along with the morphological and many other biochemical alterations during metacyclogenesis,

one major change is the lengthening of LPG molecules on the surface of metacyclic promastigote

stage. It is the modified LPG that is not capable of binding to the epithelial wall of the midgut

and due to this forward migration becomes possible (Spath and Beverley, 2001; Vanier-Santos et

al., 2002). Figure 1.5 shows the structure and chemical composition of procyclic and metacyclic

Page 34: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

12

LPG of L. major (A) and the species-specific variability in the LPG of L. major, L. donovani and

L. tropica and structure of other major glycoconjugates.

Figure 1.4. Difference between the LPG of procyclics and metacyclics (A) and species-specific

variability in structure of LPG structure of other major glycoconjugates (B). {(A) from Spath et

al., (2001) and (B) from Kamhavi, (2006)}.

1.8. Genetics of Leishmania

The haploid genome of Leishmania contains 32.4–34 Mb with 34-36 chromosomes respectively

in the L. (Leishmania) and L. (Viannia) genera. The reduction in the number of chromosomes of

the latter by 1 is due to the fusion of chromosomes 20 and 34 in the former. Recent genome

sequence analyses have shown ~8,000 protein forming genes arranged mostly in single promotor

operated polycistrons of about 200 genes each known as directional gene clusters (DGCs). As a

result, post-transcriptional modification is basic in the regulation of gene expression (Llanes et

al., 2015; Marques et al., 2015; Romano et al., 2014). Of the total identified genes, Leishmania

shares ~6,000 genes with the other trypanosomatids and 95 % of these are said to be conserved

(remained unchanged) among the species for which total genome sequences are available. So far

difference of only ~2-4 hundred genes have been found among the three species L. major, L.

infantum and L. braziliensis (Fiebig et al., 2015; Peacock et al., 2007).

Page 35: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

13

Table 1.2. Summary of the genome analysis for five different Leishmania species (Llanes et

al., 2015).

1.9. Immunology of Leishmaniasis

The immune response to Leishmania infection is complex and not yet completely deciphered. It

is mediated by several different and inter-dependent players and pathways of both the innate and

adaptive or acquired prongs of the immune system. Murine models have been and are still the

main source for the in vivo investigation of immunity to Leishmania species-which, of course,

shows species-specificity on the part of both host as well as the pathogen (Lohoff et al., 1998).

1.9.1. Promastigotes in the Inoculum vs Innate Immunity

Not all the metacyclic promastigotes of the inoculum survive but rather a huge number, almost

90 % in case of L. amazonensis in murine model, suffer from complement lysis and phagocyte

killing through toll like receptors (TLRs) at the site of the inoculation. Although dendritic cells

(DC) play their part in the phagocytic killing, neutrophils are the first (30 seconds post

inoculation of L. major in C57BL/6 mice) players of innate defense that encounter the

promastigotes of the inoculum using TLRs. It has been proposed that infected neutrophils,

Page 36: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

14

especially the apoptotic neutrophils, even help the parasite to evade being killed and are carried

harmlessly to the macrophages. In other words, the promastigotes stopover in the neutrophils

(Soong, 2008; Horta et. al., 2012; Zamora-Chimal et al., 2016). A small number of resistant

promastigotes are opsonized by C3b and are phagocytosed by DC through CR1 and CR3

receptors. Phagocytosis in this way causes the production of IL-2 in the DC and the inactivation

of protein kinase C and Mitogen activated protein kinase (MAPK) which is subsequently

preventive to oxidative burst and macrophage activation (Zamora-Chimal et al., 2016).

1.9.2. Anti-Inflammatory and Immunomodulatory Effect of Sand Fly Saliva

There have been several direct and indirect evidences that factors in the sand fly saliva pose anti-

inflammatory effects by modulation of the immune response. Saliva has been shown to contain a

variety of pharmacologically active substances including immunomodulatory proteins. These

modulators mostly cause impaired antigen presentation and suppression of enzymes that cause

prevention of oxidative burst by the macrophage (Andrade et al., 2007; Rogers et al., 2009;

Loría-Cervera and Andrade-Narváeza, 2014). It has been shown that factors in the saliva of both

Phlebotomus papatasi and Lutzomyia longipalpis inhibit Th-1 expressing cytokines from

macrophages and up regulate Th-2 expression. Dendritic cells are also inhibited by factors in the

saliva of P. papatasi and P. doboscqi, and probably Lutzomyia longipalpis, from antigen

presentation through autocrine signals in the form of prostaglandin E2 and IL-10 that act by

down regulating the MHC-II on cell surface. The same cytokines of the dendritic cells also

inhibit and diminish the infiltration to the site of inoculation through the inhibition of neutrophil

chemotactic factors MIP-1α, TNF-α and Leukotriene B4 (Abdeladhim et al., 2014;

Selvapandiyan et al., 2014). Sand fly saliva has recently got prominence in the development of

immune protection against leishmaniasis and the vaccine design. This has been inspired by

induction of antibodies against sand fly saliva and the development of protection in animal

model pre-exposed to the bite of uninfected sandflies or vaccinated with either whole saliva or

purified salivary proteins. Anti-saliva antibodies IgG1, IgG4 and IgE has been demonstrated in

the serum of both the experimental models as well as the human volunteers from non-endemic

foci exposed to experimental bite of non-infected Lutzomyia longipalpis. Patients from endemic

areas of visceral leishmaniasis when were investigated had higher levels of IgG (Bandrade et al.,

2007; Bezerril et al., 2012; Loría-Cervera and Andrade-Narváeza, 2014). Immune response to

Page 37: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

15

sand fly saliva is, however, variable with respect to species. As evaluated in the mouse model,

mice pre-immunized with the saliva of Lutzomyia intermedia did not cause protection against L.

braziliensis infection while a protective response was achieved with the saliva of L. whatmani in

the form of INF-γ producing lymphocytes (Gomes et al., 2016).

1.9.3. Host Immune Evasion

One of the important properties of Leishmania parasite is its mechanism to evade the host

immune response by modulating it. The evasion starts from innate response which is the earliest

to encounter the parasite at the entry site and leads to the adaptive one. This is achieved by

altering the function of both dendritic cells (DCs) and macrophage. The functionally altered DCs

produce insufficient amount of pro-inflammatory cytokines including INF-γ, α and IL-12 and

produce anti-inflammatory molecules like TGF-β and IL-10. Due to this, macrophage on the

other hand is prevented to produce oxidative burst which help Leishmania parasite establishing

inside its phagolysosomes (Gannavaram et al., 2016).

Surface molecules of metacyclic, not procyclic, promastigotes, especially LPG, GP63, GIP and

GIPL, play important role in subversion of host immune response and ensuring parasite survival.

Apart from these surface molecules, a variety of secreted molecules and enzymes including

cysteine peptidases, β-mercaptoetahnol dependent metalloproteases, acid phosphatases and

proteophosphoglycans of amastigotes affect macrophage anti-parasite mechanism. In Leishmania

major metacyclic promastigotes, LPG prevents the membrane attack complex by inhibiting the

conjugation of C5 and C9 to subvert complement lysis. Similarly, in case of both L. major and L.

amazonensis, gp63 cleaves C3b to C3bi and opsonization with C3bi causes internalization

through complement receptor CR3 rather than CR1 through C3b. This prevents respiratory burst

as mentioned before. It is LPG that delays the phagosome-endosome fusion by suppression of

the endosomal markers rab7 and LAMP-1 and the accumulation of F-actin. LPG and gp63 also

provide protection against the hydrolytic enzymes of the lysosome. LPG provides strong

negative charge and galactose-manose repeating units that prevent the enzyme attack. GP63

activity at acidic pH of the phagolysosome shows its function against the proteolytic enzymes

but is not yet certain as mutants can also survive. In vitro studies reveal that macrophages

incubated with LPG of GIPL of Leishmania did not express inducible nitric oxide synthase

(iNOS) upon stimulation by INF-γ and/or lipopolysaccharides (LPS). This is a necessary enzyme

Page 38: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

16

to produce nitric oxide, a component of the oxidative burst. Rather newly found molecules of

Leishmania the peroxidoxins LcPxn1 and 2 and the superoxide dismutase scavenge the nitrites

and reactive oxygen intermediates to prevent respiratory burst (Olivier et al., 2005). Toll like

receptors (TLRs) of the macrophage recognize the LPG ligand on metacyclic and amastigote-

specific antigens. This initiate a protective immune response by inducing IL-12, TNF-α and

reactive oxygen species. As investigated in Leishmania major and L. donovani, however, the

parasite has evolved mechanism to subvert the response. In the former, protein molecules SOCS-

1 and SOCS-3 signals down regulate the TLR2 using cytokines while in the latter host

deubiquitinating enzyme A20 is induced to impair the TLR2 mediated immune response. L.

amazonensis encounter the TLR2 response by producing the double stranded RNA dependent

protein kinase and IFN-β. TNF-β upregulate the production of superoxide dismutase that

subverts oxidative burst (Gupta et al., 2013)

Recently, role of the purinergic pathway in the immune evasion by the Leishmania parasite has

been described by various studies and has been comprehensively reviewed by de Figueiredo et

al. (2016). ATP release during cell injury causes inflammation by activating macrophages and

DCs through P2X7 receptors. To control inflammation and tissue damage, immune cells degrade

ATP to ADP and then AMP through ecto-nucleoside triphosphate diphosphohdrolase (E-

NTPDase) or CD39. AMP is then degraded to phosphate and adenosine through ecto-5´-

nucleotidase (CD73). Normally, through transporters, the adenosine concentration is kept

equimolar on both sides of the membrane. In case of infection or injury, however, extracellular

concentration of adenosine reaches to maximum of 10 μM from normally in nM. This

extraordinarily raised concentration is attributed to have role in upregulating IL-10 and

suppressing nitric oxide. Along with other trypanosomatids and Legionella, Leishmania species

especially L. tropic and L. amazonensis have been shown to express these enzymes and to

enhance IL-10 production and consequent NO suppression by the dendritic cells and

macrophage. Presence of AMP, adenosine, pyrases and 5′-nucleotidase in the sand fly saliva and

their role in anticoagulation by prevention of platelet aggregation and in immune modulation

have been confirmed in various studies. Figure 1.6 summarizes the immune evasion mechanism

and survival in the macrophage.

Page 39: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

17

Figure 1.5. Immunomodulation mechanism in macrophages (Bogdan and Röllinghoff, 1998).

1.9.4. The Oxidative Burst

During oxidative burst, macrophage releases reactive oxygen and nitrogen species (ROS and

RNS). Most important of the former includes superoxide anion (O2ˉ) while nitric oxide (NO) and

NO2 in the latter. Super oxides are induced spontaneously upon internalization of the parasite but

to produce nitric oxide macrophage stimulation by INF-γ and tumor necrosis factor (TNF) is

necessary. Production of NO occurs due to the reduction of N-terminus of L-arginine by

inducible nitric oxide synthase (iNOS or NOS2) the synthesis is NADPH mediated and occurs

upon stimulation by INF-γ and TNF (Horta et al., 2014; Carneiro et al., 2016). ROS not only

kills the parasite but also cause apoptotic killing of host cell by targeting the enzymes of signal

transduction especially the protein-tyrosine phosphatases (PTPs) that control vital physiological

functions. Killing of the infected cells by the immune system enhance the chances of pathogens

Page 40: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

18

clearance. Target site of ROSs in the PTPs is rich cystine residue. Leishmania parasite however

has evolved mechanism to counteract the cleavage of PTPs by ROS. L. donovani has been shown

in vitro in murine cell line RAW 264.7 to subvert the oxidative burst killing and apoptosis of

host cells as well as itself. The subversion has been found to be suppression of cytokine signaling

(SOCS) mediated thioredoxin induced healing of PTPs by reduction. In this way parasite can

maintain the environment that provide tenancy for it (Srivastav et al., 2014).

1.9.5. Activation of Natural Killer Cells (NK) and Adaptive Immunity

Activation of natural killer cells (NK) is initiated in the lymph nodes by the solubilized LPG

from the complement lysed promastigotes and LPG presented in association with the CD86, TLR

and MHC-II receptors by the DCs that have overcome and killed phagocytosis resistant

promastigotes. This activate the NK cells and T cells thereby initiating adaptive response. Thus,

the initial acquired or adaptive immune response to Leishmania, like many other intracellular

pathogens, is, as evaluated in murine models, through the cell mediated prong of the acquired

immunity. Thenceforth, the humoral response is initiated through the recruitment of specific

subtypes of helper T-cells. Th 1 response is inhibitory to the infection while Th 2 response

favours the establishment of infection (Schleicher et al., 2008; Zamora-Chimal et al., 2016;

Zamora-Chimal et al., 2017).

1.9.6. Role of Natural Killer Cells

Natural killer cells are lymphocytes that are from 5-20% of the total lymphocytes in the blood.

These are classified as CD3ˉ and CD16/56+. There are two functional subsets i.e., cytokine

producing ones and cytotoxic ones. Apart from the above-mentioned activation mechanism,

direct contact of the promastigotes with NK cells can also contribute to their activation (Leike et

al., 2011). NK cells are the primary source of INF-γ in the early days of infection which is

necessary for oxidative killing of the parasite by macrophage and for the priming of T-cell

especially the protective Th1 response (Glennie and Scott, 2016) that also produce INF-γ but

several weeks later (Scharton and Scott, 1993). Figure 1.6 shows the mechanism of how NK

cells are activated and what they do?

Page 41: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

19

Existing interpretation of the stimulation and role of NK cells in experimental visceral leishmaniasis (i.v. injection

of mice with L. donovani). Metacyclic Leishmania promastigotes are internalized by CD11c+ myeloid DC, that

produce the NK cell- triggering cytokine IL-12 in a TLR 9-reliant way (Schleicher et al., 2007). Furthermore, to IL-

12 the cytokine IL-2 is vital for the initial instigation of NK cells, while IL-18 plays a contributing character and IL-

15 is totally expendable in this procedure. Acquaintance of plasmacytoid dendritic cells (pDC) to L. promastigotes

centrals to a huge release of type I interferons (IFN-α/β), which, is of slight significance for the start of splenic NK

cells. Infected macrophages (MΦ) are unaffected by NK cell lysis in vitro and in vivo, however, NK cells produce

IFN-γ, that triggers MΦ for killing amastigotes, a practice that needs TNF and iNOS articulated by the MΦ.

Figure 1.6. Activation and role of NK cells in Experimental Visceral leishmaniasis (Bogdan et

al., 2012).

Page 42: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

20

1.9.7. Th1 and Th2 Response-Mechanism and Outcome

Susceptibility or resistance of host is strongly associated with the immune response that is

triggered by the Leishmania parasite. The response itself is influenced by a variety of host and

parasite related factors including species of the parasite, its genetic structure and host genetic,

biochemical and physiological standing. As investigated in the mouse and other animal models

of cutaneous and visceral leishmaniasis (Figure 1.7) and as described before Th1 response leads

to parasite clearance and healing while Th2 causes the establishment of parasite and its

persistence in the host cells. The two responses present different cytokine profiles and a

consequent variable recruitment of the macrophages. Dominant cytokine of the Th 1 response are

INF-γ, interleukins 1, 2 and 12 and TNF-α. During the Th 2 response interleukins 4,5,10 and 13

and TGF-β are produced that orchestrate a different mechanism of macrophage activation. Apart

from these, IL-27 and IL-17 are also important for launching an inflammatory response (Ajdary

et al., 2009; de Santana et al., 2017).

Figure 1.7. Immune response in cutaneous leishmaniasis infection (Abdossamadi et al., 2016).

Page 43: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

21

Defensive character of IL-17 against intracellular amastigotes in visceral leishmaniasis has been

demonstrated by comparing its level in patients and controls. A higher level of IL-17 in the

patients before treatment is decreased but still higher than the control can be attributed to its role

in the orchestration of protective immune response. It has been demonstrated to synergize with

interferon gamma and elicit NO production by the MΦ (Savoia, 2015). Similarly, a mixed type of

Th1 and Th 2 immune response in chronic and none-healing patients infected with L. tropica has

been recognized. It is attributed to the production of cytokines of the two responses including

IFN-γ, IL-13 and IL-5 (Ajdary et al., 2009) as well as the less described cytokines CD26 and

CD30 (Ajdary et al., 2007).

1.10. Treatment Approaches for Leishmaniasis

Being a neglected disease and causing a 60,000 annual death toll and annual morbidity of 2,090

thousand disability-adjusted life years, leishmaniasis is one of the major tropical diseases that

needs a special concern regarding drug development. It is because till date no reliable vaccine is

available against the disease (Evans and Kedziersky, 2012; Brikholtz et al., 2011) and the

available chemotherapy, the mainstay to encounter the disease, has lost its effectivity against

several, if not all, strains of the Leishmania parasite. The main factor in the chemotherapeutic

failure is development of resistance by the parasite strains especially in the endemic foci for the

various forms of the disease e.g., visceral leishmaniasis in North Bihar (India) and Sudan (Ponte-

Sucre et al., 2017). In addition, the first line of defense against the parasite including pentavalent

antimonials, the polyene anti-fungal amphotericin B and its liposomal preparation (AmBisome)

and antibiotic pentamidine and drugs passing the clinical trials have a variety of other limitations

(Seifert and Croft, 2006). High cost, side effects and hospitalization for treatment are the main

categories of limitations of the drugs in market (Nagle et al., 2014). Research regarding drug

discovery is in progress for both visceral and cutaneous forms of leishmaniasis. Cutaneous

leishmaniasis is especially on the list of Drug for Neglected disease initiative (DNDi) that is a

non-profit organization for drug development and research for neglected diseases (Mears et al.,

2015). A variety of synthetic and natural organic molecules and inorganic compounds are being

tested for their bioactivity against leishmaniases. Active drug targets in the parasite proteome are

of special concern and are under extensive investigations. Immunomodulant are being searched

both in the natural and synthetic pool of chemical compounds. Phyto materials have, however,

Page 44: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

22

been the primary source of not only folkloric medicine for two millennia but also provide us with

39% of the currently available therapies in the market (da Rosa et al., 2017).

The Genomic Institute of the Novartis Foundation (GNF) is one of the leading bodies aiming at

the drug research and development against kinetoplastids. After screening 700,000 small

molecules against the extracellular bloodstream stages of Trypanosoma cruzi and axenic

amastigotes of L. donovani, 2,000 hits (which are different molecules having revealed inhibitory

effects) have been identified. Out of these hits, 44 % have also shown antipathogen activity

against the intracellular stages of T. cruzi. While such attempts have not been as much

productive in case of Leishmania as for T. cruzi, high-throughput image-based screening assays

by automated confocal microscopy against intracellular form have dug out 350 hits out of

300,000 screened. Regarding promastigote stage, only 4% of the hits active were also found

active against intracellular amastigote stage. A more advanced and efficient approach to screen

molecules against intracellular pathogens might be ex vivo cultures of infected target organs with

genetically modified fluorescent strains. Such has been of significance in case of luciferase

containing L. donovani in murine splenic explants that could identify 200 active hits out of 4,035

compounds. The same is also under extensive investigation using L. major, another important

species of the pathogen. Alongside this, half a million different compounds have been passed

through preclinical trials in in vivo murine models using fluorescent and bioluminescent

protocols to monitor parasite load in relation to dose of candidate compounds (Ang et al., 2015).

Since the trend to develop new drugs is rather new one instigated by realization of resistance

acquired by pathogens to the existing drugs, it will take us time to find out new therapies

especially for eukaryotic parasites that are metabolically and biochemically more diverse and

complex. Rapid developments in the proteomics and genomics are certainly of importance in the

drug development and innovation. It is because they are unravelling hidden biochemical and

metabolic mysteries of pathogens and provide potential targets for compound screening. Certain

computational techniques (a multidisciplinary approach requiring contributions from

mathematicians, chemists and chemical engineers, parasitologists, pharmacists and computer

scientists) are playing magnificent role in predicting pharmaceutical potential of organic and

inorganic molecules (Sinha et al., 2017).

Page 45: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

23

1.10.1. Antimonials

Antimonials, as mentioned before, have been first line of defense against leishmaniasis for about

7 decades and continue to be the primary treatment available in most of the countries where the

disease is endemic. As trivalent sodium antimonial tartrate, they were first used in 1905 against

trypanosomes by Plimmer and Thompson. As antileishmanial, trivalent antimonials were first

used against cutaneous leishmaniasis by Vianna and against visceral leishmaniasis by Di Cristina

and Caronia in Sicily and Rogers in India in 1915. At that time the drugs were, however, found

highly toxic due to which the use was almost abandoned. It was in 1925 when urea stibamine, a

pentavalent form, was synthesized and used to save many lives in India by Brahmachari. It took

another 20 years to develop one of the currently used antimonials pentavalent sodium

stibogluconate (Pentostam) in 1945 via an intermediate antimony gluconate (Solustibosan) in

1937 (Nagle et al., 2014).

Instead of known resistance developed by the parasite against the drug, high toxicity

(pancreatitis, hepatitis and liver enzyme dysfunction, arrhythmias, muscle and joint pains),

lengthy therapy and poor tolerance, we are compelled to still use it due to the unavailability of

affordable and safe alternate chemotherapy. Currently a 20-28-day therapy at 20mg/kg/day is

recommended by WHO for cutaneous leishmaniasis that has shown species specific and

geographic limitations. A 30 to 60 % failure rate has been noted in case of L. (V.) braziliensis

and 30 % treatment failure against L. (V.) guyanensis (Zauli-Nascimento et al., 2010). Due to

increased acquired resistance, the cure rate in India (the endemic focus of Northern Bihar) for

VL is less than 50% (Sundar and Singh, 2016). Although in use since 1905 in various forms, the

resistance mechanism as well as the mechanism of action of antimonials is still not completely

known. What we know in this respect, has also been inferred from experiments primarily done

with arsenate. To grasp the Leishmania amastigotes, a drug has to pass through diverse physical

barricades e.g., the plasma membrane of macrophage, parasitophorous vacuole and the parasite

membrane. This is the most immediate obstacle that affects the success of drugs against

intracellular parasites. Resistance to the antimonials might be due to the parasite-specific

mechanism of reduction of active form of drug in the intracellular environment. This might

probably be achieved by a variety of different mechanisms including low uptake, enzymatic

inactivation and metabolism of drug, efflux of drug and sequestration in cell organelles. Most of

Page 46: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

24

the information regarding the resistance acquisition has, however, been inferred indirectly by

using pentavalent arsenate, a related compound, either in vitro or in murine models using limited

number of parasite species (Ashutosh et al., 2007; Yadav et al., 2015).

As such both forms of the drug are accumulated at pharmacological concentrations in both the

stages of various species of Leishmania, SbV shows negligible antileishmanial activity. It is now

almost a fact that the trivalent SbIII, and not the pentavalent prodrug SbV, is the active form

against the parasite. It has been suggested the pentavalent form is first reduced to the trivalent

form inside the parasite. The reduction has been speculated to be enzymatic through mediation of

both the host and parasite thiols or non-enzymatic where thiols themselves may bring

conversion. The parasite enzymes involved are thought to be thiol-dependent reductase or

antimoniate reductase or both. The parasite derived thiols that might mediate the conversion are

thought to be glutathione-spermine conjugate and trypanothione. Host derived thiols including

glutathione (macrophage cytosol), cysteine and cysteinyl-glycine (lysosome) are proposed to be

the main reducing agents in this process. Among these the latter two and trypanothione of the

parasite that work at lower pH have been found more efficient (Ashutosh et al., 2007; Frézard et

al., 2009).

The uptake of antimonials, especially the SbV, is not all clear. As evaluated in axenic

amastigotes, SbV uses protein receptors that resemble that of gluconate because gluconate

compete with SbV. The non-competition of pentavalent arsenic and phosphate for SbV rules out

the proposition that it uses transporter of these. Since both the forms of the drug accumulate

differentially in the axenic amastigote, it is logical to say that both the forms use different

mechanism to enter the parasite. As evaluated by the uptake experiments using both trivalent

arsenic and antimoniate, these use GlpF (an aquaglyceroporin that facilitate the entry of glycerol

in E. coli). These are also facilitated by aquaglyceroporins in the yeast and mammalian cells

(Brochu et al., 2003; Mandal et al., 2015).

1.10.2. Amphotericin B

Amphotericin B is a naturally occurring antimycotic that belongs to the polyene (heptaene)

subgroup of macrolides (Hamilton-Miller, 1973) obtained from Streptomyces nodosus. It is

strongly lipophilic and in conjugation with liposomes presents highly changed pharmacokinetic

properties than the conventional form; the amphotericin B deoxycholate (D-AMB). The

Page 47: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

25

conventional form of the drug manifests severe infusion reactions like fever, chills and

thromboblephitis and cause renal toxicity, lowering of blood potassium level and myocarditis.

Although lipid formulations do not lack these complexities but present these with comparatively

less intensity and severity. Lipid based formulations being less toxic and more efficacious are of

great concern in the current scenario of drug development (Silva-Jardim et al., 2014; Yousuf et

al., 2016; Kip et al., 2017). Three main lipid formulations currently approved by FDA are

liposomal amphotericin B (LAMB), amphotericin B lipid complex (ABLC) and amphotericin B

colloidal dispersion (ABCD) (Hamil, 2013) respectively known by their trade names

AmBisome®, Abelcet ® and Amphocel TM or Amphotec ® (Kip et al., 2017). Amongst these,

AmBisome have been the most extensively used in clinical trials and in the treatment of visceral

leishmaniasis. In comparison to other liposomal formulations, AmBisome has been found more

effective and less toxic in mouse model. It was found more effective in reducing burden of

Aspergillus fumigatus in experimentally infected mice than Lambin®. Toxicity of AmBisone

was comparatively less than Lambin®. It has also been found less toxic and more effective than

another liposomal formulation Anfogrn in the treatment of experimental fungal infections in

mice (Azanza et al., 2015).

Normally used as second-line of therapy against VL and aggravated cases of CL and MCL,

amphotericin B is used as primary cure of VL in Bihar (India), where L. donovani has become

resistant to antimonials and have been abandoned (Croft and Oliaro, 2011; Singh et al., 2017).

FDA approved dose for LAMB against VL (Savovia, 2015) is 3 mg/kg/day for

immunocompetent patients on days 1-5, 14 and 21 while that for immunocompromised

individuals a dose of 3-5 mg/kg/day on days 1-5, 10, 17, 24, 31, and 38. More than 97% cure rate

has been achieved with this regimen (Georgiadous et al., 2015). However, for reasons apparently

unclear, variable degree of efficacy of liposomal formulations of AMB have been reported in

other studies. The variability in efficacy might be attributed to administration regimen, clinical

form of leishmaniasis as well as geography and ethnicity. Balasegaram et al. (2012) reviewed

and shared expert opinion regarding the use of LAMB (AmBisome) against various forms of

leishmaniasis. Their study revealed that the drug and its various formulations has mainly been

used against visceral leishmaniasis. They found scarce and limited record of amphotericin B use

against cutaneous and mucocutaneous leishmaniases. They have reported a cure rate of 95% and

98% with AmBisome at a single dose of 10 mg/kg in phase-III and 20 mg/kg over four doses in

Page 48: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

26

phase-IV clinical trials respectively in South Asia, Latin America and Europe. This regimen has,

however, given poor results in East Africa. In combination with 100 mg/kg miltefosine for 8

days and 15 mg/kg parmomycin for 11 days, 5 mg/kg AmBisome has given a cure rate of 97.5%

each in South Asia.

Amphotericin B targets ergosterol, a lipid that is shared by fungi and Leishmania species in their

cell wall and cell membrane respectively. After binding to the ergosterol, the complex forms a

transmembrane channel that turns the membrane permeable to cations, water and glucose. The

impaired permeability leads to altered membrane potential due to the loss of potassium ion and

consequently cell death occurs (Saha et al., 1986; Ordóñez-Gutiérrez, 2007; Ayres et al., 2008).

Some of the recent studies have also investigated the interaction of amphotericin B with the host

macrophage cholesterol and impairment of promastigote-macrophage attachment and subsequent

internalization (Paila et al., 2010). While this is the case, we can suppose that loss of ions and

impairment of permeability of host cells, especially muscle cells, when AMB interacts with the

membrane cholesterol can be attributed to the toxicity of the drug on the host.

1.10.3. Pentamidine

It is an aromatic diamidine and primarily an antibacterial agent. It was first implied in the

treatment of visceral leishmaniasis as well as the various forms of cutaneous leishmaniasis in

1950s in India and Spain. It was however, in use for the treatment of trypanosomes since 1940s.

Due to higher degree of toxicity it is usually used as second-line of chemotherapy where the

antimonials fail to show efficacy (Loure and Yorke, 1939; Croft and Coombs, 2003; Nagle et al.,

2014; Griensven et al., 2016). In French Guiana and Suriname, it is used as first-line of treatment

for CL with dosage regimen of 3 mg/kg over 4 intramuscular (IM) injections administered every

second day or a single IM dose of 7 mg/kg. The latter might be repeated after 48 hours if needed.

With the former regimen, a cure rate of 35 % against cutaneous infection of L. braziliensis in

Peru and 96 % for CL in Colombia have been achieved as compared to the standard antimonial

therapy. With the latter dosage regimen, 78.8% and 83.6% of the CL patients could be cured.

Apart from this, other studies in Colombia has reported 95% cure rate in the case of CL. In case

of VL, however, a cure rate of 67-77% was acquired with a 15-injections course of 4 mg/kg/day

administered over 14 to 30 days in 1990s. Other studies have reported 98% cure rate with the

standard dose of 4 mg/kg/day three times a week in India. Mediterranean VL caused by L.

Page 49: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

27

infantum have been treated successfully with 2-4 mg/kg/day over 3-5 weeks treatment. After

this, in concern of rapid acquisition of resistance, its use was abandoned due to switching to

amphotericin B deoxycholate as second line drug. When it comes to toxicity, its use is limited

because it causes insulin-dependent and irreversible diabetes mellites with an incidence rate of 4-

12%. Other complications associated to its use include nephrotoxicity, myocarditis, pyrexia,

hypoglycemia and hypotension (Balaña-Fouce et al., 1998; Mitropoulos et al., 2010; Nagle et

al., 2014). A recent study used pentamidine as prophylactic agent to prevent relapse in

immunocompromised patients over 12 months duration. For this purpose, they employed a single

dose of 3-4 mg/kg body weight once in 3-4 weeks. With this prophylactic regimen, relapse

prevention was 79% and 71% after 6 and 12 months respectively (Diro et al., 2015).

1.10.4. Miltefosine

The general class to which miltefosine belongs is alkyl phosphocholine which are basically

ether-alcohols. Its IUPAC name is hexadecyl 2-(trimethyl-azaniumyl) ethyl phosphate and is also

known as hexadecylophosphocholine. Its empirical formula is C21H46NO4P with molecular

weight of 407.57 g/mol. Other related compounds used as drugs are edelfosine, ilmofosine and

perifosine. Miltefosine was originally developed as anti-neoplastic agent and was used in topical

formulations for the treatment of tumors of breast by two different independent groups in

Germany and United Kingdom in 1980s. Later, its antileishmanial activity was found in vitro as

well as in vivo studies. Currently it is a licensed oral antileishmanial agent in India against L.

donovani (Dorlo et al., 2012). A comprehensive review of the chemistry and pharmacology of

the drug as anticancer and antileishmanial agent has been presented in the reference quoted

ahead. Miltefosine (Impavido) is administered orally for 28 days at 2.5 mg/kg body weight. With

this regimen a cure rate of 94-97% have been successfully achieved in India against Kala-azar

(Eissa et al., 2012; de Morais-Teixeira et al., 2011). This has, however, dropped to 84 % in India

and Bangladesh in recent years. The cure rate against cutaneous infection caused by L.

panamensisis and L. braziliensis in Colombia was once more than 90 % but has gradually

reduced to 69.8 % in case of the former and to 49 % in the latter case. Studies in Brazil have

reported 75 % and 71% cure rate against the cutaneous disease respectively caused by L.

braziliensis and L. guyanensis. In Bolivia against the mucocutaneous infection by L. braziliensis

in Guatemala against the cutaneous leishmaniasis caused by L. braziliensis and L. mexicana and

Page 50: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

28

in Afghanistan against L. tropica, a cure rate of 70 %, 53 % and 63 % have been achieved

respectively (Verla et al., 2012).

The miltefosine family of drugs cause apoptosis of Leishmania parasite, most expectedly by

direct degradation of the parasite DNA (Khademvatan et al., 2011; Verla et al., 2012).

1.10.5. Paromomycin

Paromomycin or aminosidine is an aminoglycoside antibiotic metabolite of Streptomyces

rimosus (Pujol-Burgués et al., 2014) that has been known and used as antileishmanial over the

last 60 years. However, its use as antileishmanial agent has been scattered and seemingly less

frequent than antimonials in the past. While it was found more efficacious in antimony resistant

cases during various clinical trials of both the VL and CL, it has been approved for parenteral

administration especially in the endemic state of Bihar in India where antimony resistant L.

donovani is prevalent. Reluctance in the use of paromomycin through parenteral route is due to it

being hepatotoxic, ototoxic and causing pain at the site of administration (Croft et al., 2006;

Banerjee et al., 2011). It is a WHO recommended first-line drug in combination with sodium

stibogluconate (SSG) against VL in Sudan. It is usually administered parenterally over 17 days

as compared to the 30 days regimen of SSG alone. The regimen is followed for both the freshly

infected and SSG treated relapse cases (Atia et al., 2015). Experimental topical ointments of the

aminosidine over two weeks treatment have been found to effectively reduce the parasite burden

and achieve clinical cure of CL. A more prolonged four-week topical application have been

effective in curing 2/3 of the patients in Iran leaving 1/3 to get antimonial therapy (Asilian et al.,

2003). Liposomal formulations for both the topical and parenteral use have almost traveled a

successful journey through the lab dish and animal trials to reach the clinical use (Bavarsad et

al., 2012). In conjugation to the liposomes, paromomycin is better absorbed, well tolerated and is

with improved therapeutic index (Gaspar et al., 2015).

1.10.6. Phytomedicine in Experimental and Clinical Trials

As mentioned before, plants have been, are and most certainly will be the primary source to

serve human with remedies for his illnesses. Many unexplored compounds in plants can act as

effective, harmless remedies and as replacement of the existing medications (Kaur et al., 2014).

The bulk of recent literature presenting experimental and trial-based use of thousands of plant-

Page 51: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

29

derived agents against various species of Leishmania in vitro and in vivo in animal models is, of

course, hard to review completely. However, it is an attempt to review at least a fraction of the

scattered wealth of knowledge regarding phytomedicine in various infections including CL.

Plant derived compounds act against pathogens either directly by physically damaging them,

targeting their biosynthetic pathways and by modulating their immune systems. Withania

somnifera or winter cherry also known as Indian Ginseng, Asgand and Koti Laal in Pushto

language have been a candidate in folk remedies throughout the Sub-continent. It has been

extensively investigated for bioactivities and have revealed antioxidant, anticancer, anti-

inflammatory, anti-rheumatic, anti-tuberculosis, antianxiety, senescence preventive and

immunomodulatory properties. Withaferin-A and Sitoindosides VII-X isolated from this plant

have been associated with the above-mentioned activities and proliferative effects on superoxide

dismutase and catalase. Its constituents have been shown to positively affect leukopoiesis. W.

somnifera extract is hepato and nephro-protective and has anti-leishmanial activity exerted

through immunomodulation has recently been explored. A dose of 200 mg/kg in combination

with Aspharagus racemosus extract significantly reduced burden of L. donovani in liver and

spleen of BALB/c mice and induced strong Th 1 response. As evaluated by liver and renal

function tests, animals treated with the herbal extract had normal values as compared to the

control and SSG treated groups. At a dose of 350 mg/kg along with 5 mg/kg cisplatin, having

known hepatic and renal toxicity, significantly minimized the toxicity and enhanced parasite

clearance. However, when administered alone showed less potency against the pathogen. This

substantiated its role in immunomodulation. At 200 mg/kg along with A. racemosus extract and

at 350 mg/kg alone, it positively regulated the Th 1 cytokines (IFN-γ, IL-2, IG 2a) and antibodies

and downregulated those of the Th 2 cytokines including IL-4, IL-10 and IG1 (Sachdeva et al.,

2013; Alali et al., 2014; Kaur et al., 2014). W. somnfera constituents withanolides especially

chemotypes NMITLI-118, NMITLI-101R and withaferin-A have caused strong protective

immune response against L. donovani in hamster. In the treated groups, mRNAs protective

immune mechanism including inducible nitric oxide synthase, IFN-γ, IL-2 and TNF-α

heightened significantly but a reduction was seen in the anti-inflammatory cytokines like IL-4,

IL-10 and TGF-β and enhanced Leishmania-specific LTT response and elevated IgG2, ROS and

NO were seen. Intensity of these activities was more in NMITLI-10 followed by NMITLI-118

and withaferin-A (Tripathi et al., 2014). When administered to experimentally infected hamsters

Page 52: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

30

in combination with the ED50 doses of miltefosine (10 mg/kg), paromomycin (30 mg/kg) and

amphotericin B (0.5 mg/kg) for 5 days each, the chemotype NMITLI-101R caused 98%, 94%

and 93% clearance respectively. This also exerted some effects on immunomodulation. It had a

strong effect on the up-regulation of inducible nitric oxide synthase and Th1 cytokines like IL-12

and IFN-γ while the Th2 cytokines IL-4, IL-10 and TGF-β were down regulated. This therapy

was also effective in generation of reactive oxygen species, nitric oxide, Leishmania-specific

IgG2 and significantly enhanced the delayed type hypersensitivity (Tripathi et al., 2017). In vitro

assays of crude extracts of the W. somnifera against the promastigotes and amastigotes of L.

donovani have been found effective with IC50 of 78 and 63 μg/mL. Withaferin-A, a constituent

isolated from the crude extract was found highly active against both promastigotes and

amastigotes with the IC50 values of 12.5 and 9.5 μg/mL respectively (Sharma et al., 2009).

Essential oils from a variety of different plants have been the most used plant derived bioactive

agents showing a wide range of activity against bacteria, fungi, viruses, and protozoa.

Commercial use of 300 of the currently known 3000 plant derived essential oils in the

pharmaceutical, agronomy, food, cosmetic and sanitary industries is on the record. Main

constituents of many of these oils are terpenes and terpenoids and other aromatic compounds.

Terpenes in these oils are mono-, di- and triterpenes. Monoterpenes are, however, in the highest

quantity (up to 90 %). Most of such oils contain a few components in higher concentrations

while the rest are present only in fractions (Akhtar et al., 2014; Le et al., 2017). Chenopodium

ambrosioides has been used over centuries as anti-parasitic remedy in South American folk

medicine. Main component of the plant that have been used and have potency against helminths

and protozoans is its essential oil. The essential oil is further constituted of three principle

components, carvacrol (62%), ascaridole (22%), and caryophyllene oxide (5%). These oils have

been commercially extracted and used as anti-parasitic agents but at a stage were abandoned to

use due to toxicities and sometime fatalities both in human and experimental animals. It is

genotoxic and causes gastroenteritis characterized by immediate hyperemia, leading to nervous

symptoms like headache, facial flushing, impaired vision, vertigo, incoordination and paresthesia

(Mnzote et al., 2009). Antimicrobial and anti-parasitic activity of the oil is extensively being

investigated. It has shown good in vitro activity against a variety of Gram positive and Gram

negative bacteria and different strains of Candida albicans (Brahim et al., 2015). The oil has

Page 53: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

31

shown promising results, both in vitro and in vivo in mouse model, against L. amazonensis. In

vitro 50% effective dose of 3.7 and 4.6 μL/mL have been achieved against the promastigotes and

amastigotes respectively. When administered at 30 mg/kg orally, intraperitonially and

intralesionally, this essential oil showed variable degree of antileishmanial activity against L.

amazonensis. It was more effective in reducing parasite burden intraperitonially followed by oral

and intralesional treatment. Intraperitoneal administration was, however, more toxic than the

others (Monzote et al., 2006; Monzote et al., 2007). In another experiment, when administered at

a dose of 30 mg/kg every day for 14 days intralesionally brought significant healing and parasite

clearance in the footpad lesions of experimentally infected BALB/c mice with L. amazonensis.

As against this, an artificial mixture of main constituents i.e., ascarodole, carvacrol and

caryophyllene oxide at the same dose regimen in 30:70 DMSO-saline solution was not found

effective and caused mortality. Lesion morphology and morphometry in this case was

comparable to that treated with the mixture of DMSO-saline and the untreated control. Positive

control treated with 28 mg/kg glucantime caused less healing than the essential oils (Monzote et

al., 2014). Hydroethanolic extract of C. ambrosioides administered intralesionally caused a

significant reduction in the parasite load of L. amazonensis in footpad lesions, draining lymph

nodes and spleen in relation to standard antimony treatment and negative control. Treatment

regimen followed was 5 mg/kg extract and 28 mg/kg meglumine antimoniate every 4th day for 5

treatments. Oral treatment did not show satisfactory results in the parasite reduction. Heightened

production of nitric oxide was observed in the culture of peritoneal and lymph node macrophages

showing immunomodulatory effect of the plant (Patrício et al., 2008). Different combination of

the three major constituents of C. ambrosioides essential oil gave variable results against L.

amazonensis both in vitro and in vivo in BALB/c mice. A 1:4 ascaridol-carvacrol combination

gave excellent results in vitro against both the promastigotes and intracellular amastigotes and

was not cytotoxic. The same combination in a 20:80 mg/kg ascaridol-carvacrol was found far

better than the control in reducing the lesion size and the parasite burden (Pastor et al., 2015).

The action mechanism of ascaridol has been evaluated in the promastigotes of Leishmania

tarentolae. Once inside the parasite, it is cleaved in the presence of ferrous ion to release radicals

that are toxic to the parasite (Geroldinger et al., 2017).

Page 54: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

32

Syzigium cumini (Jamun) essential oil with 87.7% monoterpenes has shown in vitro

antileishmanial activity against L. amazonensis promastigotes. An IC50 of 36 mg/L was obtained

at 24 hours incubation of promastigotes with the oils (Dias et al., 2013). A purified constituent of

S. cumini essential oil α-pinene, has shown promising results against both the stages of L.

amazonensis. IC50 values of 19.7, 16.1 and 15.6 μg/mL have been respectively achieved against

the promastigotes, axenic amastigotes and intramacrophage amastigotes. It was found to exert

the antileishmanial effect through enhancing phagocytic and lysosomal activity of the

macrophage and raising production of nitric oxide. They presented negligible cytotoxicity as

evaluated by haemolysis and exposure to the murine macrophages. The half minimal cytotoxic

concentration (CC50) and half minimal haemolytic concentration (HC50) of the essential oil and

α-pinene were 614.1 and 425.2 μg/mL and 874.3 and 233.3 μg/mL respectively (Rodrigues et al.,

2015). When tested against the same species of the parasite, its hexanoic extract showed

considerable leishmanicidal activity against the promastigote giving an IC50 of 31.64 μg/mL

(Ribeiro et al., 2014).

Demarchi et al. (2015) extracted and used essential oils and 6,7-dehydroroyleanone from

African plant Tetradenia riparia (ginger bush) against promastigotes and intramacrophage

amastigotes of L. amazonensis. Incubation of the promastigotes with the compounds for 24, 48

and 72 hours gave the LD50 values of 0.5, 0.3 and 0.8 μg/mL and 16.9, 14.9 and 3 μg/mL for the

essential oil and 6,7-dehydroroyleanone respectively. Morphological alterations were also

induced in the treated promastigotes. Both the agents caused a significant reduction in the

infection index in the murine macrophages infected with amastigotes and caused little

haemolysis in human erythrocytes. The compound 6,7-dehydroroyleanone was, however, toxic

to murine macrophages.

Thirty-seven essential oils from Vietnamese plants were tested against promastigotes of L.

mexicana at 25 and 50 nL/mL by Le et al., (2017). Of 37 oils they found 5 oils with considerable

leishmanicidal activity with 99% promastigote killing at the 25 nL/mL and an IC50 value of less

than 10 nL/mL. These 5 oils were from Amomum aromaticum (IC50=9.25 nL/mL),

Cinnamomum cassia (IC50=2.92 nL/mL), Elsholtzia ciliate (IC50=8.49 nL/mL), Ocimum

gratissimum (IC50=4.85 nL/mL) and Zingiber zerumbet (IC50=3.34 nL/mL). When evaluated

Page 55: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

33

for cytotoxicity against WI-38 fibroblasts and J774 macrophages, no toxicity was reported. This

study provided valuable preliminary information about the bioactivity and cytotoxicity of the 5

plants. Further studies were suggested to work out the lowering of cytotoxicity and efficacy in

the animal models and beyond.

Currently, literature regarding use of phytochemicals against microorganism in general and

Leishmania in specific is growing like weeds, therefore bulk of literature available on the use of

plant extracts, fractions, and isolated compounds against Leishmania is practically not possible to

review here. Over the last three decades, extensive work has been done throughout the world

both in developed and developing countries and it would need a book of several volumes to

completely accommodate the data. Although most, if not all of the phytoproducts are used in the

lab dish and against animal model stages of Leishmania. The hugeness of literature can be

illustrated with a simple example of a review published by Soosaraei et al. (2017), where7500

articles were found from 1999 to 2015 on electronic databases like EBSCO, ScienceDirect,

PubMed, Google Scholar, Scopus and four Persian databases Magiran, Iran doc, Iran Medex and

the Scientific Information Database (SID). Out of these articles 1750 were found irrelevant by

title. Since they were interested in the articles published in Iran, so they included only 68 in their

review. Interestingly, in these 68 articles a total of 188 experiments using 98 plants were

performed of which 140 were in vitro and 48 were in vivo.

1.10.7. Inorganic Agents in Experimental and Clinical Trials

Over the last three decades, like phytomedicinal chemists, the inorganic and organometallic

chemists have been equally busy in developing metal-based therapies against different parasites

including Leishmania. The primary design strategies they have been focusing on are; enhancing

of the antiparasitic efficacy by the incorporation of metal centre to existing agents, improvement

of bioavailability of the existing agents by bioactive ligand coordination, targeting DNA with

metal coordinated intercalating agents, and description of metal inhibitors of parasite specific

enzymes.

Discovering group-specific common targets in Leishmania and their cousins like

trypanosomatids and subsequent development of therapeutic agents against common targets is

Page 56: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

34

like killing two birds with one stone (Gambino and Otero, 2017). Organometallic complexes of

various transition metals like zinc, copper, lead, osmium, rubidium, ruthenium, platinum,

bismuth, gold, silver, tin, nickel, boron, and cobalt have been employed for their antiparasitic

potential. Copper and zinc complexes derived from dipyridophenazine, a DNA intercalator, have

shown excellent leishmanicidal activity against both the promastigote and intracellular

amastigote form of L. infantum. The in vitro assays have, however, found them cytotoxic to

human macrophages (Madureira et al., 2013; Tahghighi, 2014). Cytoplasmic disorganization,

vacuolation, and induction of binucleated cells have been the effects of these compounds in

promastigotes (Novarro et al., 2003). Copper conjugates with β-diketonates have been evaluated

against the promastigotes of L. amazonensis. Cu(acetylacetone)2, Cu(trifluoroacetylacetone)2,

Cu(hexaluoroacetylacetone)2 and of Cu(nitrilotriacetic acid)2 gave ED50 values after 24, 48, and

72 hours of incubation as 31,84 and 94 μM, 68, 57 and 43 μM, 135, 124 and 68 μM and 263, 293

and 122 μM respectively. With the same incubation regimen, however, the ED50 values were 83,

19, 15 μM, 74, 31, 24 μM, 137, 39, 12 μM and 342, 137,13 μM respectively (Portas et al., 2012).

Lead complexes derived from 2,5-anhydro-D-manitol are potential antitrypanosomatid enzyme

inhibitors. Out of the 55 derivatives, 13 and 16 caused more than 50% inhibition of

Trypanosoma brucei phosphofructokinase and Leishmania mexicana pyruvate kinase

respectively (Nowicki et al., 2008). Three different complexes of ruthenium showed promising

activity against L. amazonensis, L. braziliensis and L. mexicana. Moreover, murine macrophages

RAW 246.7 treated with the complexes at the same concentration showed 90% viability (Costa

et al., 2017). Silver complexes of polypyridyl have been found effective against the

promastigotes of L. mexicana at 10 μM/L in vitro. [Ag (1,10-phenantroline-5,6-dione)2] NO3 and

[Ag (dipyrido[3,2-a: 2’,3’-c]phenazine)2] NO3 were shown as DNA metallo-intercalators and

were proposed to exert their antileishmanial activity through DNA inhibition (Navarro et al.,

2006). Carboxylate derivatives of tributyltin (IV) complexes have shown promising activity

against promastigotes of Leishmania tropica KWH23 at a concentration of 20 μg/mL. All the

tested compounds caused 95-97% lethality and gave 50% inhibitory concentration from 0.008-

0.954 μg/mL. Moreover, they gave much higher IC50 values when tested against the HepG2 and

THP1 cell lines at the same concentration (Waseem et al., 2017).

Page 57: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

35

Organometallic complexes of the platinum-II and ruthenium-I have been found to cause growth

inhibition of both the flagellated and amastigote stages of L. donovani. RhI(CO)2 Cl (2-

Aminobenzothiazole) complex interacted with the nucleic acid as well as protein biosynthetic

pathways of the parasite and was most potent against the parasite. It was, however, toxic to J774

murine cell line. Complexes [(Cis-PtII (DDH) (2,5-Dihidroxibenzensulfonic)2 and RhI (CO)2

Cl(5-Cl-2-Methilbenzothiazole)] gave rather unsatisfactory results causing morphological

alterations only but were found safe and nonaggressive to the J774 cells (Mesa-Valle et al.,

1996). Some complexes derived from (2,2′:6′,2′′-terpyridine) platinum-(II) have been able to

cause effective inhibition of the intracellular forms of both the L. donovani and Trypanosoma

cruzi at 1μM concentration and the extracellular form of T. brucei at 0.03 μM (Lowe et al.,

1999). Although it got fame much latter in the last quarter of 20th century with the discovery of

an organometallic complex of Platinum II, the cisplatin, an anticancer agent, scattered use of the

inorganic metals and their complexes has been known since ancient times. Important step in the

use of inorganic therapeutics was the discovery of arsenophenyglycine and arshphenamine by

Paul Elhrich for sleeping sickness and syphilis respectively (Caballero et al., 2015). Cisplatin in

vitro antileishmanial outcome has earlier been established. Kaur et al. (2010) studied

antileishmanial action of 0.5mg/kg body weight and 1mg/kg body weight of the cisplatin drug L.

donovani infected BALB/c mice. Significant reduction in the parasite burden was noted

by1mg/kg body weight. Akhtari et al. (2019) reported effective in vitro antileishmanial action of

a cisplatin nano-formulation against promastigotes and amastigotes of Leishmania major. The

IC50 of cisplatin nano-formulation was 0.11±0.09μM for amastigotes of Leishmania major i.e.,

41-fold lesser than that of Glucantime (4.52±1.31μM), signifying that a minor quantity of

cisplatin nano-formulation in contrast with Glucantime is needed to execute 50% of intracellular

amastigotes.

A complete review of the ongoing extensive investigational research on the metal-based

remedies for the parasitic diseases and physiological disorders is beyond the scope of this

document.

1.10.8. Diterpenoids

Diterpenoids is a class of natural metabolites of bacterial, fungal and plant origin widely used in

industry and medicine. They are all derived through enzymatic cyclization of (E,E,E)-

Page 58: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

36

geranylgeranyl diphosphate. The enzyme used in diterpene production is diterpene synthase

(Smanski et al., 2012). Two important sub-groups of the diterpenoids are ent-kauranes (e.g.,

steviol) and ent-beyeranes (e.g., isosteviol). Interconversion and chemical modification of these

to modify their biological and biochemical properties can be done. These have been found to

possess bactericidal, malaricidal, antidiabetic, anti-inflammatory and anti-tumour activities

(Cherney et al., 2013; Wang et al., 2018). Ent-kaurane diterpenoids ent-kaur-16-en-19-oic acid

and ent-kauran-16α-ol (Da Costa et al., 1996) and pimarane diterpenoid pimaric acid (Rubio et

al., 2005) have been investigated to effectively inhibit the growth of Trypanosoma cruzi at 500

μg/mL, 200 μg/mL and 150 μg/mL respectively. When tested against different pathogenic

protozoa, ent-3-a-hydroxy-kaur-16-en-18-ol (ent-kaurane) and ent-7-oxo-pimara-8,15-diene-18-

ol (ent-pimarane) were found much effective. The former showed significant activity against

Plasmodium falciparum (IC50=3.5 μM) and Leishmania donovani (IC50=2.5 μM) while the

latter was found effective against P. falciparum only (IC50=3.8 μM) (Nogueira et al., 2016). Iso-

kaurenoate, an esterified derivative of kaurenoic acid, showed high effectivity against P.

falciparum strain W2 (IC50=8.8 μM) (Batista et al., 2013). Members of thiosemicarbazones,

derivatives of kaurenoic acid which is a diterpenoid, have been found to possess trypanocide

activity against the Trypanosoma cruzi with IC50 ranging from 2-24 μM (Haraguchi et al.,

2011). Flavonoids from Stevia satureiifolia var. satureiifolia, also producing steviosides, steviols

and isosteviols, have been found to inhibit the epimatigotes, amastigotes and trypomastigotes of

T. cruzi and promatigotes of Leishmania braziliensis. Compounds eupatorin and 5-

desmethylsinensetin caused considerable inhibition on the epimatigotes (IC50s 0.2 and 0.4

μg/mL respectively) and trypomastigote (IC50s 61.8 and 71.1 μg/mL respectively). Compound

5-desmethylsinensetin showed IC50 of 87.7 and 37 μg/mL respectively against the amastigotes

of T. cuzi and promatigotes of L. braziliensis (Beer et al., 2016). 4-nitro- and 2,4-

dinitrophenylhydrazone, isosteviol derivative, have been found to show activity against T. cruzi

with IC50 value of 17.5 to 22.58 μg/mL (Ullah et al., 2016). Ullah et al. (2019) have

comprehensively reviewed the biological activities and synthesis of isosteviol from Stevia

rebaudiana.

1.10.9. Actinomycins

Actinomycins are the secondary metabolites of actimomycetes, a hallmark group of bacteria

regarding antibiotic production. Genus Streptomyces is of special concern and produces 35-50

Page 59: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

37

kinds of actinomycins (Katz and Pugh, 1960). Actinomycin D has been found to hamper DNA

synthesis (Cooper and Braverman, 1977), cause cytostasis and enzymogenesis (Grand et al.,

1972; Nsi-Emvo and Raul, 1984) and up to 2005 about 3300 publications it has been a subject of

(Koba and Konopa, 2005). This inhibition on DNA synthesis attributes the anti-tumor effects of

actinomycicn D and C3 (Hollstein, 1974).

1.11. Leishmania Strain to be Used

Although there had been scattered studies and reports about leishmaniasis (Rowland et al., 1999)

in Khyber Pakhtunkhwa (KP) after its emergence due to post Russian invasion and refugees’

influx to Pakistan, it was not until 2010 that a serious and detailed study about genetic

characterization of the causative agent was undertaken. Dr Nazma Habib Khab, Assistant

Professor Department of Zoology, University of Peshawar Pakistan, then PHD student at London

School of Hygiene and Tropical Medicine worked out the molecular characterization and

phylogeny of isolates from KP, Pakistan (Khan et al., 2016). Being a strain of local origin and

easily available in local repositories, Leishmania strain KWH23 was used in this tudy.

Aim and Objectives of the Present Study

This study aimed to explore antileishmanial properties of some plant and microbial metabolites.

Main objectives of this study were to;

1. Study growth characteristics and progression of Leishmania tropica KWH23 in vitro

2. Explore antileishmanial effects of Actinomycin D, Z3, Z5 and Hydrazine derivatives of

Isosteviol

3. Assess cytotoxic effects of the mentioned test compounds on human lymphocytes.

4. Evaluate genototoxic effects of the test compounds through Comet Assay.

Page 60: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

38

Chapter 2. Morphology and Culture Characterization of

Leishmania tropica KWH23

2.1. Materials and Methods

2.1.1. Culture of Leishmania tropica KWH23 Promastigotes

Leishmania tropica KWH23 promastigotes culture was initiated in RPMI-1640 (with 20 mM

HEPES and L-glutamine without NaHCO3) medium (Sigma-Aldrich Europe) at a densiy of

1×105/mL in tissue culture flask with surface area of 25 cm2, canted neck, and non-vented cap.

The RPMI-1640 medium was supplimented with 10 % heat-inactivated fetal bovine serum (hi-

FBS, Sigma-Aldrich Europe), 100 µg/mL streptomycin and 100 IU penicillin (Sigma-Aldrich

Europe). The growth was monitored daily by counting in improved Newbaur haemocytometer

over a week. The coverslip was moisturized with clean water and coverslip was affixed to

the hemocytometer. The occurrence of Newton's refraction rings below the coverslip specified

appropriate adhesion. Promastigotes were kept in in a Petri dish containing formaldehyde

impregnated tissue and all 4 sets of 16 corners were counted in hemocytometer (Figure 2.1).

These cells were averaged and multiplied by 104 to get number of promastigotes/mL. The growth

curve for Leishmania tropica KWH23 promastigotes was then made as shown in Figure 2.2.

Figure 2.1. Hemocytometer illustration representing set of 16 squares (marked in red colour)

used for counting promastigotes.

Page 61: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

39

2.1.2. Evaluation of Morphological Variations of Promastigotes during Culture

Morphologic variability was monitored daily through Giemsa stained smear over a period of one

week. About 20µL of culture was smeared on albuminized slides and fixed in methanol for 1-3

minutes after air drying at room temperature. Various concentrations of the methanol were tested

to fix the Leishmania tropica KWH23 promastigotes. Fixed smears were stained for 14-19

minutes with 1 in 10 Gibco-KaryoMAX Giemsa stain (Thermo-Fisher, Germany) stock solution

in water (WHO, 2016). Smears were observed under the powerful oil immersion objective

(1000x magnification) of the light microscope (Leitz Dialux 22 EB, Germany) and photographs

were obtained by using USB microscope camera or sometime mobile phone camera. Though

comparable in appearance and aroma to (drinking) ethanol, methanol is poisonous to the nervous

system. Contact to methanol through ingestion, inhalation, or absorption can cause impaired

vision, unconsciousness and death. Personal protective equipment (PPE) like covers for eyes

(safety glasses), face (mask) and skin (gloves) were used during fixation process.

Temporal variation in the length, width, and shape of the promastigotes and length of the flagella

were qualitatively accessed to determine different Leishmania tropica promastigote morphotypes

in the axenic culture. Metacyclic promastigotes were also assesed through scanning electron

microscopy. For the morphological examination, Leishmania tropica promastigotes were fixed in

2.5% glutaraldehyde in 0.1 M sodium cacodylate buffer for one to three hours. The coverslips

were washed in 1% HCL in industrial methylated spirit (IMS) at 50oC, washed thoroughly under

tap water, dried and stored. Afterward, promastigotes were stick to poly-L-lysine-covered

coverslips and dehydrated in ascending concentration (20%, 30%, 50%, 70% and 90%) of

ethanol, with each step of 5-10 minutes duration.

2.1.3. Cultivation of Axenic Amastigotes and Evaluation of Morphology

Axenically grown Leishmania tropica amastigotes has become an interesting life cycle stage for

drugs screening and for investigating biochemical as well as molecular variability between the

promastigote and amastigote stages. A variety of different protocols have been followed to

axenically transform promastigotes to amastigotes for different Leishmania species. In the

present study, 1x106/mL Leishmania tropica promastigotes in the stationary phase were grown at

37oC in humidified conditions lacking CO2 in RPMI 1640 medium fortified with 20% heat-

inactivated fetal bovine serum (hi-FBS, Sigma-Aldrich Europe), 100 µg/mL streptomycin, 100

Page 62: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

40

IU penicillin and L-glutamine. The pH of the medium was adjusted at 4.4 using phosphoric acid

(85% aqueous solution BDH Chemicals, UK). The amastigotes were stained with Giemsa stain,

and phase contrast microscopy was used to monitor the transformation on daily basis and fully

transformed amastigotes were also confirmed through scanning electron microscopy. During the

promastigote to amastigote transformation, the change in cell shape was observed daily by smear

fixation and staining and expressed in terms of percentage from elongated promastigote form in

the initial inoculum to the fully formed amastigotes with a variety of oval forms in between.

Presence, length and absence of the flagellum was also recorded. Reduction in the cell size was

verified through stage and ocular micrometry. The size measurement was mostly based on the

fully round and oval amastigotes. The diameter was confirmed after taking mean for 20

randomly selected individual amastigotes cell measurements.

2.1.4. Acquisition of Axenic Amastigotes by Starvation

Amastigote stages were also acquired by Leishmania tropica in the in vitro culture, when the

promastigote culture was kept starved without medium changes, accidentally for the first time,

and then several times when repeated. This was a total accidental happening, which occurred due

to a week vacation when, to be on the safe side, the culture was sub-passaged in 25 mL complete

RPMI1640 medium in 25 cm2 non-vented flasks at 26°C, the normal temperature for

promastigotes. After opening, to my surprise, the culture medium was plasma-yellow and on

observation revealed a bulk of amastigotes. These were processed as mentioned before. This

practice was then repeated several times deliberately to confirm the conversion.

2.1.5. Viability during Transformation

Viability of cells is a necessary aspect in the drug sensitivity and biological studies. The viability

of Leishmania tropica during promastigote to amastigote transformation was checked daily over

a week using trypan blue exclusion and was expressed as percentage of viable cells. After

complete transformation, the amastigotes were cultured in the normal RPMI1640 as mentioned

above for promastigotes to check the viability.

Page 63: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

41

2.2. Results

2.2.1. Growth Curve of L. tropica KWH23 Promastigotes

Flagellum, kinetoplast and nucleus are distinct morphological indicators of Kinetoplastida and L.

tropica owns a single flagellum, nucleus and kinetoplast and each duplicate once during the

entire cell cycle (Wheeler et al., 2011). The flagellum growth inducts first, trailed by mitosis

then kinetoplast division that twitches after the commencement of nuclear anaphase. The growth

of L. tropica KWH23 promastigotes through growth curve over one week period in the culture is

shown in figure 2.1. For the first three days of the culture, a stunted growth was observed. On

day 4, the culture reached log phase of the growth. On day 5th to 6th the mid and late log phases

were achieved. This means peaked growth was achieved on day 6. On day 7, the culture

inwarded stationary phase with a promastigote density of 1×107/mL. The promastigotes became

sluggish with decreased motility and were slithering around. Cells were looking stumpy with

long flagella. The promastigote body looked like a carrot with swollen anterior and tapered

posterior (Figure 2.3-b).

Figure 2.2. Representative growth curve of Leishmania tropica KWH23 in vitro.

-2000000

0

2000000

4000000

6000000

8000000

10000000

12000000

14000000

16000000

1 2 3 4 5 6 7 8

Nu

mb

er o

f P

rom

asti

gote

s/m

L

Days

Page 64: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

42

2.2.2. Axenic Promastigote Morphotypes

Various morphotypes were determined through one week developmental cycle for L. tropica

KWH23 promastigotes. Culture was initiated from stationary phase promastigotes. It was infinite

passage number of the L. tropica promastigotes strain KWH23. Promastigotes of L. tropica at

different developmental forms were observed in these 8 days, some promastigote cells were in

the course of division i.e., in mitotic (M) stage of the cell cycle, whereas the rest were at other

stages of interphase including G1, S and G2 phases. In present study a promastigote cell in M

phase was demarcated as having two nuclei and/or kinetoplasts having incomplete cytokinesis,

as noted by inspection of Giemsa-stained slides (Figure 2.3).

Figure 2.3. Different stages of L. tropica KWH23 promastigotes representing cell cycle in

culture (see text for description). A naturally occurring amastigote (Arrow in “e”) can be seen in

a 13-day old promastigote culture.

Page 65: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

43

On day one almost all of the promastigotes were nectomonads (Figure 3.2-b) having long,

slender and progressively tapering bodies with long flagella about one and a half to twice as long

as the promastigote body. On day 2 only 5% of the promastigotes were leptomonads (Fig 2.3-c)

with still slender and long cells and longer flagella almost indistinguishable from those of

nectomonads. In leptomonads, the cell body tapered posteriorly and cell to flagellum ratio

decreased. On day three, the ratio of leptomonads rose to 44 %. During the log phase on day 4,

metacyclics appeared in the culture. The matacyclics to leptomonads to nectomonads were 27%,

43% and 29% respectively. Metacyclics had long flagella and short stumpy cell bodies with

anterior end much broader than the posterior as compared to that in leptomonads and

nectomonads. The posterior end of the body was bearing a slender tail like extension (Figure 2.3

d-f). Promastigotes number increased till day 6 ( late log phase), where onward the stationary

phase reached and the number of metacyclics increased progressively. Following the

development of the parasite in the present study, the procyclic promastigotes were enormously

infrequent (Figure 2.3-a) and infrequently oval amastigotes were also observed in Giemsa

stained smears (Figure 2.3-e).

2.2.3. Division of L. tropica Promastiogtes in Axenic Development

The nucleus, kinetoplast and flagellum provide distinct morphological indicators. A typical L.

tropica promastigote cell have single kinetoplast, nucleus, and flagellum and each of which

duplicates once during the cell cycle (Wheeler et al., 2011). During logarithmic growth, 17% of

the promastigotes were found dividing, so different features of the dividing cells were recorded

(Figure 2.4). Most of the individuals duplicated the flagellum first following division of the

kinetoplast and the nucleus respectively. However, in rare cases, division of the kinetoplast and

nucleus preceded flagellar splitting (Figure 2.4 C, D, and E).

Wheeler et al. (2011) reported interesting events through phase contrast and fluorescence

microscopy in L. mexicana promastigote cell cycle. They presented new flagellar emergence

from the flagellar pocket at 5.2-hour, DNA segregation at 6.3-hour (mitosis) and finally the

kinetoplast partition at 6.6-hour time period respectively. We also observed “rosettes” which are

bigger bunches of cells placed side-to-side with flagella in the direction of the centre of the

cluster. Rosettes are also involved in a rare swarming behaviour, flocking into prolonged

Page 66: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

44

clusters. Iovannisci et al. (2010) showed rosettes as an unrecognized stage in the life cycle of

Leishmania and hypothesized that rosettes initiate mating in Leishmania.

Figure 2.4. Division of the L. tropica promastigotes in axenic development. Cells with 2 flagella

single kinetoplast and single nucleus (A and B), Cells with single flagellum, two kinetoplasts and

single nucleus (C), Cells with Single flagellum, two kinetoplasts and two nuclei (D and E) and

Cells with two flagella, two kinetoplasts and two nuclei near to the completion of cytokinesis

(F). Solid arrows show kinetoplasts and the others indicate flagella.

2.2.4. Cultivation of Axenic Amastigotes

After 24 hours of incubation under the above-mentioned conditions, the metacyclic

promastigotes started to change in the overall morphology. In transformation from promastigote

to amastigote stage, major changes have been observed regarding the morphological changes in

the cell shape and length of flagellum (Figure 2.5).

Page 67: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

45

Figure 2.5. Promastigote to amastigote transformation. Change in the cell morphology and loss

of flagellum is seen progressively.

2.2.5. Changes in Outline and Flagella during Differentiation of Promastigotes to

Amastigotes

Leishmania experiences differentiation from sandfly promastigote form into intramacrophage

amastigotes, a progression that is vital for its intracellular existence. The changes in shape of the

cell and length of the flagellum during promastigote to amastigote transformation/ differentiation

were documented (Table 2.1). Leishmania are exposed to pH changes through their life-cycle.

Sandfly blood meal contains procyclic promastigotes which reproduce in the pH 7.3-7.4 of the

bloodmeal, while amastigotes are adopted to acidic environment of the macrophage

phagolysosomes (pH 3.2-5.5). Acidification of phagosome is critical defensive mechanism in

phagocytes activating microbial killing.

We observed amastigote stages when Leishmania tropica in the in vitro promastigote culture was

kept starved without fresh medium changes, by chance for the first time, and then after several

Page 68: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

46

times when repeated purposely. This was a total accidental happening, which occurred due to

one-week holidays and when, on the safe side, the culture was sub-passaged in 25 mL complete

RPMI-1640 growth medium in 25cm2 non-vented flasks at 26°C, the normal temperature for

promastigotes. The results are presented as percentage of various forms (morphotypes) in 100

cells counted in a Giemsa stained smears. During 10 days period, daily monitoring of the

transformation (differentiation) processes displayed dissimilar morphotypes. There was a steady

loss of the flagella with individual variability. As indicated in table 2.1, interestingly, on day 1,

dissimilar morphotypes were observed and till day 3 a mixture of morphotypes was present. Day

4 and onward only the rounded forms with no flagella and oval forms with no flagella were

observed. However, few rounded forms with scarcely visible flagella were also present in the

culture. Although the transformation process started right on the following day of culture in

starvation (conditioned/used) medium, however, proper amastigote conversion was observed

after day 4 onwards. The fully transformed culture contained the round and oval forms.

However, oval cells predominated the rounded ones throughout the culture of amastigotes of L.

tropica isolated from CL lesions from the type locality of the strain (Figure 2.5).

Cell size as well as length of flagellum experienced tremendous reduction during the

transformation of Leishmania tropica KWH23 promastigotes to amastigotes. There was

significant reduction (10.97 to 4.51µm) in cell length on day one, moderate reduction on day two

(4.51 to 3.93µm) and day three (3.93-3.28µm) in acidified medium respectively. We measured

pH of the medium and interestingly it was from 5.0 to 5.5 and in this acidified medium

(temperature 26°C and pH 5.5) transformation/differentiation was satisfactory. From day four till

day seven the length of the amastigote forms became stable measuring between 3.19 to 3.11µm

respectively (Table 2.2). Fully transformed amastigotes varied from little oval to near rounded

(intermediates) to fully rounded (Table 2.3). The oval amastigotes measured 3.65±0.36 µm in

length and 2.94±0.28 µm in width, while the complete rounded forms were between 3.18±0.31

µm to 3.15±0.30 µm in diameter on day 5 and day 10 in conditioned medium respectively.

Page 69: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

47

Table 2.1. Promastigote to amastigote transformation ratio over 10-day duration based on

Leishmania morphology. *****

*****RNF=Rounded no flagellum, RBVF=Rounded barely visible flagellum, RSF=Rounded short flagellum,

RLF=Rounded long flagellum, ONF=Ovoid no flagellum, OBVF=Ovoid barely visible flagellum, OSF=Ovoid short

flagellum, OLF=Ovoid long flagellum, ENF=Elongated no flagellum, EBVF=Elongated barely visible flagellum,

ESF=Elongated short flagellum, ELF=Elongated long flagellum,

Shape

Variation→

Day

RNF RBVF RSF RLF ONF OBVF OSF OLF ENF EBVF ESF ELF

1. 03 05 05 02 04 25 27 09 08 05 04 03

2. 10 09 05 01 23 27 20 02 0 0 02 01

3. 09 08 07 01 31 40 03 01 0 0 0 0

4. 19 13 0 0 68 0 0 0 0 0 0 0

5. 31 06 0 0 63 0 0 0 0 0 0 0

6. 29 09 0 0 62 0 0 0 0 0 0 0

7. 41 0 0 0 59 0 0 0 0 0 0 0

8. 44 0 0 0 56 0 0 0 0 0 0 0

9. 47 0 0 0 53 0 0 0 0 0 0 0

10. 43 0 0 0 57 0 0 0 0 0 0 0

Page 70: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

48

Table 2.2. Size distribution of various differentiating and transforming Leishmania tropica

KWH23 amastigotes in acidified medium.

Days in

Culture

Mean Diameter in µm Variance Standard

Deviation

Confidence

Interval (CI) of

the Variance

Standard Deviation of

the Confidence Interval

0 10.97 2.36 1.54 1.4-5.4 1.2-2.1

1 4.51 0.70 0.84 0.4-1.7 1.18-1.45

2 3.93 0.29 0.54 0.17-0.70 0.413-0.840

3 3.28 0.06 0.25 0.036-0.15 0.189-0.387

4 3.11 0.04 0.21 0.025-0.10 0.158-0.316

5 3.19 0.06 0.25 0.035-0.138 0.187-0.371

6 3.11 0.04 0.21 0.025-0.10 0.158-0.316

7 3.14 0.05 0.23 0.030-0.118 0.173-0.343

Table 2.3. Size distribution of the completely round, intermediates and oval forms of

Leishmania tropica amastigotes.

Fully Rounded Amastigotes

Mean

Diameter

in µm

Variance Standard

Deviation

Confidence Interval

(CI) of the Variance

Standard Deviation

of the Confidence

Interval

Day-5 3.18 0.1017 0.32 0.05-0.25 0.224-0.50

Day-10 3.15 0.095 0.31 0.048-0.22 0.219-0.47

Oval Amastigotes

Length 3.65 0.133 0.36 0.071-0.33 0.266-0.57

Width 2.94 0.084 0.29 0.045-0.21 0.212-0.46

Page 71: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

49

2.2.6. Viability during Transformation

Viability of cells is a necessary aspect in the drug sensitivity and biological studies. The viability

of Leishmania tropica KWH23 cells during promastigote to amastigote transformation was

checked daily during the study period using trypan blue exclusion and was expressed as

percentage of viable cells (Fig. 2.6). Leishmania tropica differentiation from promastigotes to

amastigotes stage viability was assessed by differential total of lifeless and living parasites using

the trypan blue colourant exclusion technique. Results presented in figure 2.5 corresponds to

three independent experiments performed in triplicate. Results clearly present non-significant

difference in Leishmania tropica amastigotes survival by the 0.4% trypan blue dye exclusion

technique throughout the experimental time (day zero to day 7). We observed maximum

survival rates of 96-98% throughout our experiments when amastigotes were exposed to various

available nutrients in acidified complete growth medium at 37oC.

Figure 2.6. Viability assessment for Leishmania tropica amastigotes during transformation and

differentiation.

0

10

20

30

40

50

60

70

80

90

100

0 1 2 3 4 5 6 7

% V

iab

ilit

y

Days

Page 72: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

50

2.2.7. Natural Acquisition of Axenic Amastigotes

During the study period, when the culture was kept starved for one week to ten days at 26⁰C and

in the absence of O2, the promastigotes changed to amastigote stages due to change in pH of

medium (from pH 7.4 to 5.0). This occurrence was purely unintentional for the first time, during

which colour of the RPMI 1640 medium changed from pink to light yellow with pH of 4.8-5.0

(Figure 2.6). The complete culture medium, in the repeat experiments, having 10% HI-FCS

started changing its colour after day 3 with pH falling to 6.8 having only promastigotes. On day

5 the pH dropped to 6.0 but it still was having only promastigotes in late log phase. The pH fell

gradually to 5.5 on day 7 with 82% promastigotes and the rest were partially changed

(intermediates) amastigotes having rounded to oval or stumpy body but long flagellum. On day 9

the pH further dropped to 4.9 to 5.0 and the percentage of amastigotes increased to 80% mostly

having rounded body and diminutive to rudimentary flagellum. On day 10 onwards the pH

dropped to 4.8 and the proportion of amastigotes invigorated to 91% and interestingly viability

of the amastigotes was 97% as assessed by trypan blue exclusion test. The culture had 1.2 x 107

amastigotes per mL. When grown in fresh complete medium for promastigotes at 26oC the

amastigotes successfully transformed back to promastigotes. Natural transformation and

differentiation were slower than the one performed in the defined medium for axenic

amastigotes. However, the results were satisfactory, and the amastigotes yield was enough. Such

transformation yielded enough axenic amastigotes for one-time use in an experiment. The

amastigotes could be successfully propagated through sub-passaging in the axenic amastigotes

defined medium.

C

Page 73: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

51

Figure 2.7. Unsurprisingly acquired amastigotes (A), Starved culture medium with pH 4.8-5.0

having naturally changed amastigotes (B). Standard RPMI 1640 growth medium with pink

colour and phenol red as a pH indicator (C).

2.2.8. Requirements for Counting, Fixation and Smearing of Leishmania tropica KWH23

Counting is a necessary process in culturing and propagation as well as for in vitro and in vivo

anti-leishmanial assays for testing various compounds. To count Leishmania parasites,

promastigote form needs inactivation by diluting in 2% formaldehyde in PBS, because they are

fast moving. Amastigotes are, however, stationary and need no such inactivation treatment.

During the present study, 10-25 % ethanol was found acceptable to inactivate them to be easily

studied under microscope in haemocytometer. Ten percent aqueous methanol solution for 10-30

seconds gave excellent results in fixing both the promastigote and amastigote forms of the

parasite. For adhesion of the parasite to the slide surface, egg albumin was used. Fresh album

was less effective in holding the parasite to slide surface but its room storage for one to 7 days

made it much effective in attaching the Leishmania cells to the slide surface. Hen egg albumin

contains 385 amino acids with molecular mass of 42.7 kDa. Rehault-Godbert et al. (2010)

showed that hen egg albumin contains many molecules good for human health and with many

antimicrobial proteins which inhibits growth of Salmonella enterica after its storage at 4, 20, or

37°C for 30 days preceding to inoculation. Amastigotes were found less effective in sticking to

the surface of albumenized slides than the promastigote. A 2 % (V/V) working Giemsa stain

solution for 14-20 minutes at room temperature gave excellent results in staining both

promastigotes and amastigotes. This working Giemsa stain solution functioned reasonably for 2

weeks without any granulation when placed in dark. Fresh stain replacement after two weeks,

however, is recommended.

2.2.9. Revision of Strain Nomenclature

Leishmania tropica KWH23 strain was originally isolated from a 5-year-old boy from Jamrud (an

endemic region for cutaneous leishmaniasis) in Khyber Agency of Pakistan who visited Kuwait

Teaching Hospital, Peshawar, for treatment in 2010. It was isolated by our colleague Dr. Nazma

Habib Khan during her PhD from London School of Hygiene and Tropical Medicine. Since then

it has been labelled with the name where KWH comes from the name of the hospital where the

Page 74: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

52

strain originated and 23 refers to the serial number of the patient studied visited on that day.

According to the strain nomenclature for Leishmania isolates, it must contain four parts

separated by slash (/). The first part is a reference to the host from which it is isolated. It is

usually a four-letter abbreviation. The first letter represents the class of the host in the hierarchic

classification and the following are first three letters of the genus of the host. The second part is a

two-letter abbreviation of the country of origin, a list of which is available online. The third part

is the year of isolation. The fourth part is not fixed and is on the choice of the person who has

done the isolation. It might be the original patient ID in the hospital record or of the researcher.

So as per Article 50.1 and Endorsement 50A of the International Code of Zoological

Nomenclature the name of this Leishmania tropica KWH23 strain technically be written as

MHOM/PK/2010/KWH23.

2.3. Discussion

Morphological variability of promastigote stage of different Leishmania species have been

described in the vector host. Several different Leishmania morphotypes occurs during

development in the Phlebotomus sand fly vector. Each of these morphotypes also presents

bichemical and molecular variability with diverse virulence to the host (Molyneux and Killick-

Kendrick, 1987; Schlein, 1993; Ismael et al., 1998; Gossage et al., 2003; Kamhavi, 2006;

Oliveira et al., 2009; Ramalho-Ortigao et al., 2010; Dougall et al., 2011). The description of

such morphotypes in axenic development is apparently difficult partly because of temporal

association of such developmental forms and partly due to great deal of species-specific

variations. For these reasons, limited number of studies have been attempted to assess the

morphological forms in the axenic cultivation of the Leishmania promastigotes. Such studies

have been done on limited number of Leishmania species e.g., L. donovani, L. mexicana and L.

major being the prominent ones (da Silva and Sacks, 1987; Sacks, 1989). However, axenic

growth of the promastigotes reflect several differnet morphotypes realised during the in vivo

dvelopment in the sandfly vector. Different morphotypes found in the sandfly vector are also

reported from the in vitro culture (Sacks et al., 1984; Gossage et al., 2003; Lei et al., 2010a).

Initiating from CL lesion amastigotes, Bates (1994) demonstrated, various morphotypes of L.

mexicana axencally. The initial amastigote to promastigote transformation was achieved in 2

days. Logarithmic growth, with a mixture of dissimilar morphotypes in the multiplicative stage,

Page 75: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

53

was noted to occur on days 3 and 4. At this point, very low density of the infective metacyclics

was distinguished. Most of the promastigotes on the late day 3 were longest (15.22 ± 3.49 μM)

and extremely slender than noted on any other day and were carrying long flagella. However,

flagellar and body length was comparable. These were morphologically much similar to the

nectomonads noted in in vivo studies. On day 7th , the promastigotes reached to the stationry

phase and metacyclogenesis was on its peak. In 9 days old axenic culture, however, the density

of metayclics reached to 95%. The metacyclics became shorter, were broader anteriorly and

needle sharp posteriorly and were with long flagella. The flagella were 1-2 times longer than the

cell size (7.59 ± 1.15 μM). These findings are in agood concensus with what we came across in

the present study. Short and slender metacyclic promastigotes with long flagella have been

demonstrated in stationary cultures by other investigators also (Sacks and Perkinn, 1984; Grimm

et al., 1991). The ratio of metacyclics to other forms is in inverse relationship to the number of

passages in our experiments. It usually decreases in long-term axenic cultivation. Fresh cultures

that have gone through few passages get fast metacyclogenesis with higher densities of

metacyclics than the older cultures (Cysne-Finkelstein et al., 1998). Results of the present study

are strongly supported by the in vivo results shown by Dougall et al. (2011), where similar

morphological forms of promastigotes of an unidentified species of Leishmania isolated from

day biting midges of genus Forcipomyia. They reported long slender nectomonads with extended

flagella. The leptomonads they isolated were rather short banana like with relatively short

flagella. Their metacyclics, however, varied from needle long shape with long flagella to

anteriorly broad and posteriorly needle sharp morphotypes having long flagella. Lei et al.

(2010b) also reported procyclic, nectomonad, leptomonad and metacyclic stages in Leishmania

culture. They have shown similar body lenghts for procyclics, leptomonads and metacyclics with

length ranging from 6-11 μm. However, nectomanads were more than 12 μm in length. All of the

stages they recorded have been with similar morphology to what we have found in the present

study.

Lectins obtained from differnet plants including peanut, are used to purify metacyclic

promastigotes from culture. These proteins agglutinates the procyclic and other forms leaving

metacyclics only. Metacyclics and non-metacyclics separated from stationary culture of L.

amazonensis have been reported to be morphologically different. The non-metacyclics have been

long and almost equally broad along their length except near the posterior tip. These have been

Page 76: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

54

shown to have extended flagella. These are the characteristics of the nectomonads. The

mectacyclics have been short and stumpy with broader anterior half and much narrower posterior

half. These have been shown to possess very long flagella (Saravia et al., 2005). This study

support our results. Soares et al. (2005) also noted similar results for L. braziliensis. The

procyclic form was found to be much variable in size of the cell and flagellum. They probably

considered all the non-metacyclic forms as procyclic that varied in cell size from rounded to

elongated individuals. However, the metacyclics were defined as smaller and thinner posteriorly,

with long flagella.

Bee et al. (2001) grown promastigotes of L. mexicana under different temperatures and pH and

found metacyclics with different growth and pro-amastigote transformation kinetics. At a

temperature of 26°C and pH of 7.2 no transformation was recorded. In these physiological

conditions, however, in some experiments transitional forms have been seen for short duration.

At temprature of 26°C and pH 5.5, greater degree of transformation have been observed but it

was followed by reversion to promastigotes in 24 hours. When the transformation was checked

at 32° C and pH 7.2, within 12 hours almost all of the cells entered to the intermediate forms that

sustained for 48 hours. Although they did not revert to the promastigote but did not fully

transformed. A temeperature and pH panel of 32°C and 5.5 was the most effective in bringing

the desired transformation over 48-96 hours (Bee et al., 2001). These conditions for

transformation are in close association to our observation. Similarly, the observation of various

trnasitional stages in the above mentioned study share what we noted in this study. However, we

did not see any reversion like they mentioned at the ideal combination they used. A difference

from what they have observed in our study is that we recorded good amastigote transformation at

26°C and 4.8 pH in the natural starvation medium.

Metacyclic promastigotes of L. mexicana grown in Schnider’s medium supplimented with 20%

fetal calf serum at 25°C and 5.5 pH have been successfully transformed to amastigotes by rising

simply the temperature to 32°C in the parent culture. After 24 hours of incubation at 32°C

temperature, more than 80% of the cells changed to rounded amastigotes with complete loss of

flagella. The remaining 20% were the intermediate forms differing in their cell and flagellar

morphology. By the day 2, however, 100% transformation was observed (Bates, 1994). These

findings present a strong correlation of temeterature and pH to the transformation process.

Page 77: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

55

Species-specific variability can, however, not be ruled out. Generally, a temeperature and pH

combination of 32-37°C and pH of 4.5-5.5 seems feasible for bringing satisfactory

transformation. Fully trnasformed amastigote may be ovoid to fully rounded in almost all

Leishmania species.

Morphological variability in the in vitro development of L. tropica can be determined

microscopically. The present study will provide guidelines for the subsequent workers in

acquisition of any specific morphotype for their use. This will also help them in acquiring axenic

amastigotes of the species by simply starvation of promastigotes. A comprehensive protocol for

processing the prmastigotes as well as amastigotes for both light and electron microscopy will be

of help to the future projects.

Page 78: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

56

Chapter 3.0. In vitro Cytotoxicity Assays of Actinomycins and Isoteviol

Derivatives on Human Peripheral Blood Lymphocytes

The key assays used during in vitro harmfulness estimation are called as cytotoxicity assays,

which concentrate mostly on cell death or to quantity growth weakening. Numerous assays are

established to determine in vitro toxicity such as enumerating cell survival /death by evaluating

integrity of the plasma membrane, overall cell protein content and certain vital functions.

Cytotoxicity testing is intended to assess the inherent capacity of a compound to kill the cells.

Along with dosage, toxicology of a compound depends upon the length of exposure and the

compound’s toxicity mechanism (Ferro & Doyle, 2001).

In sum, life ends in death and death in cells occur either by necrosis or by apoptosis. One of the

life's majestic sarcasms is that it also rests on death. The term necrosis originates from the

Greek core “necros” meaning ‘dead’, and denote accidental demise of cells exposed to extreme

ecological or heritably programmed insults (Walker et al., 1988). Wounded cells experiencing

necrosis show gross morphological and ultrastructural changes that differ sharply with those

displayed by cells enduring apoptosis. Necrotic cell death is escorted by swelling of the cell,

widespread plasma membrane lost integrity and endocytosis, expansion of cell organelles,

clumping and arbitrary degradation of nuclear DNA (Hall et al., 1997). Necrosis terminates with

the removal of cell corpses in the nonexistence of apparent phagocytic and lysosomal

participation (Galluzzi et al. 2018).

Programmed cell death (PCD) is cascade of inherent suicide pathways, vital for tissue

homeostasis, growth, development and pathogenesis. PCD was originally considered identical

with apoptosis; however, in recent times, alternate mechanisms of PCD have been described,

viz. necroptosis, autophagy and apoptosis. PCD can happen in the absence of any exogenous

environmental agitation, henceforth, working as a built-in effector of physiological agendas

for development and cellular turnover. Apoptosis or type I cell death exhibits cytoplasmic

contraction, plasma membrane bulging, chromatin shortening (pyknosis), nuclear

disintegration (karyorrhexis), and ending with the creation of seemingly integral small

vesicles (apoptotic bodies) that are proficiently taken up by adjacent cells by phagocytosis and

their degradation within phagolysosomes. This cascade of activities leads into cell death.

Cytotoxicity can be measured by diverse approaches like vital dyes viz. trypan blue exclusion

Page 79: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

57

testing and propidium iodide uptake assay, protease biomarkers activity and MTT assay (Fuchs

and Steller, 2015; Galluzzi et al,. 2018).

Previously performed cytotoxicity assays accompany all classes of drug sensitivity studies. For a

material to be a therapeutic one, it is essential that it must not be damaging to the host cells or at

least should have minimum cytotoxicity at the recommended dose which is bearable and

repairable by the body. Following this principal, all the test compounds and the standard drug

were subjected to prior cytotoxicity evaluation using human peripheral blood lymphocytes, an

easily available celly type for in vitro testing. These cells are the the very first nucleated cells

that come across any material, therapeutic agents for example, that enters the body through

vascular fluid, the blood.

3.1. Materials

3.1.1. Chemicals and Solutions

Ficoll polymer, RPMI-1640 growth medium, Phosphate-Buffered Saline (PBS), Trypan blue

solution (0.4%), XTT (2,3-Bis-(2-Methoxy-4-Nitro-5-Sulfophenyl)-2H-Tetrazolium-5-

Carboxanilide), Dimethyl Sulfoxide (DMSO), Heat-inactivated fetal bovine serum (hi-FBS),

Histopaque-1077 (Sigma H8889), Streptomycin-Penicillin solution (All from Sigma-Aldrich,

Europe).

3.1.2. Disposables

Centrifuge tubes (15 and 50 mL), 5-10 mL EDTA tubes, 5-20 mL syringes, 1.5-2.5 mL

Eppendorf tubes, 20-1000 μL pipette tips, Glass slides, Thermo Scientific Nunc™ Cell Culture

Treated Easy-Flasks™ vented (close cap) and filtered cap culture T25 and T75 flasks, Nunc™

96-Well Polystyrene Flat-Bottom Microwell Plates, 5-10 mL pipettes, Aluminium foil

(Thermo-fisher, Germany; Bio Tek GmBH, Germany; Sigma-Aldrich, Europe).

3.1.3. Apparatus

Thermo-Fisher Scientific Direct Heat CO2 Incubator, DAIHAN MaXterile™ Digital Fuzzy-

control Autoclave 80 Litre (Daihan, Korean), Thermo-Fisher Scientific Finn-pipette FpF3

micropipettes (2-200 and 100-1000 μL capacity), Microbiological Laminar flow hood (Technico

Scientific Supply, Lahore), Olympus CKX41 Inverted microscope (Olympus, Singapore),

Page 80: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

58

Brightfield compound microscope (Leitz, Germany), Heraeus Centrifuge Megafuge X1R

Refrigerated (Thermo-Fisher, Germany), Bio-Tek Microtiter plate reader with a branded system

(Bio Tek GmBH, Germany), Microbiological Incubator (Gallen-kamp Fistreem International

Limited, UK).

3.2. Methods

3.2.1. Isolation of Human Peripheral Blood Lymphocytes

Human peripheral blood lymphocytes (PBLs) are an easy source for in vitro cytotoxicity

experiments. In the present study, freshly isolated PBLs were used to test the cytotoxic effects of

the compounds. Briefly, fresh 5 mL blood was collected from verbally consented healthy BS and

M.S Zoology students using sterile 5 mL disposable syringe. The blood was immediately added

into 5-10 mL Di Potassium EDTA containing screw capped sterile polystyrene tubes. The blood

was mixed with PBS in 1:1 in a 15mL centrifuge tube. The diluted blood was carefully layered

up on equal amount over a 5 mL Ficoll Histopaque in two other tubes. Layering was done slowly

by tilting the Ficoll/Histopaque containing tube at an angle and pouring blood along the down

facing inner wall of the tube using an automatic pipette filler. This preparation was then

centrifuged at 400xg for 30 minutes at room temperature. The mono-nuclear cells were isolated

through a pipette. The isolated cells were washed three times in PBS at 400xg for 10 minutes

each. The cells were then incubated at 37oC for 2 hours in complete RPMI-1640 medium to

remove the monocytes by their affinity to adhere to the plastic and glass surfaces. The suspended

cells were then collected and pelleted by centrifugation. The pellet was resuspended in 1 mL

RPMI-1640 medium and the cells were counted in haemocytometer. Cells from different

individuals were processed separately. All the steps after the collection of blood were performed

aseptically in a laminar flow hood. As blood is a potential infectious material, therefore all the

disposables used were disinfected overnight in phenyl before proper disposal.

3.2.2. Inspecting Viability of Lymphocytes

The dye exclusion assessment is shaped on the idea that viable cells having intact cell

membranes do not take up colourants like trypan blue, eosin and propidium, however lifeless

cells are permeable and mesmerize the dye. Trypan blue (C34H28N6O14S4) is an azo dye i.e., having

Page 81: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

59

the colouring azo function (N=N-), which is an organic molecule containing two neighbouring

nitrogen atoms sandwiched between carbon atoms. To check the viability of the lymphocyte

cells, 10-50 µL of lymphocyte cells suspension was added to 10-50 µL 0.4% trypan blue solution

(in PBS), mixed by pipetting up and down and incubated at room temperature for 2-3 minutes.

The mixture was then placed on a glass slide and the percentage of viable cells was determined

by counting 100 cells. The percentage of viable blood lymphocytes was calculated by dividing

the number of viable cells by quantifying total cells and multiplying by 100. Viable cells were

the ones that did not stain with the trypan blue dye (clear cytoplasm) as viable membranes are

impermeable to the trypan blue while dead lymphocytes cells were with permeable membranes

having blue cytoplasm.

3.2.3. Plating Lymphocytes and Adding Compounds

Lymphocytes were dispensed in flat bottom 96 well polystyrene plate at 2×104 cells/well in

100μL complete RPMI-1640 growth medium. The trial compounds and the standard miltefosine

were applied in 0.02, 0.2, 2, 20, 25, 50, 75 and 100μM in triplicate for each concentration. A

negative control was kept with only 1% DMSO, the solvent of the test compounds and

miltefosine. The concentration of the DMSO, until mentioned otherwise, was always kept as 1%.

The lymphocytes were incubated at 37°C in the CO2 incubator for 48 hours.

3.2.4. Evaluation of Cytotoxicity and Cell Death in vitro

After 24 hours exposure to the test compounds, the lymphocytes cells were incubated with 20μL

of XTT solution for 2 hours at 37°C in the CO2 incubator. After 2 hours, the absorbance was

read in Bio-Tek ELx800 ELISA microplate reader through GEN5 software at 490 and 630 nm

wavelength. The formazan assembly by XTT reduction is soluble in water and therefore was

directly detected in lymphocytes supernatants so was easily calculated by colorimetric assay.

3.1.5. Data Analysis

The 96 well flat-bottomed plate was abstemiously shaken to dispense the orange colour evenly

and absorbance for lymphocyte cells and the blank background control (RPMI-1640 growth

medium only) wells was read at a wavelength between 450-490 and 630nm. The absorbance data

was first manipulated by subtraction of the 630nm value from 490 nm value for each well. Then

values obtained after substracting were then processed in excel and converted to percent

Page 82: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

60

inhibition by using the formula (control-treated/control×100). The percent inhibition values were

then processed by GraphPad Prism 6 software to calculate the IC50 values for the test

compounds and the standard drug miltefosine.

3.3. Results

3.3.1. Cytotoxic Impact of Miltefosine on Lymphocytes

Miltefosine (Hexadecylphosphocholine) was used as a standard positive control (as it is the only

drug use for the treatment of all form of Leishmaniasis orally) for evaluating its cytotoxicity

against human peripheral blood lymphocytes. It gave an IC50 value of 272.2 μM (95 % CI=

143.6 to 515.7 μM). In this respect, it may be considered as comparably less cytotoxic to the

lymphocytes as it inhibited less than 50% of the cells even at its highest concentration as

revealed in the figure 3.1 after incubation for 48 hours. Among the test compound, variable

degree of cytotoxicity was observed as compared to the standard miltefosine.

Miltefosine has been shown to bind peritoneal macrophages and Leishmania plasma membrane

membrane-bound proteins in large quantities via a detergent-like action and are causing

structural and organizational alterations showing noticeable increase in dynamics and exposure

to aqueous milieu, so resulting in membrane fluidity (Fernandes et al., 2017).

Page 83: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

61

Figure 3.1. Concentration dependent cytotoxicity of Miltefosine against human peripheral blood

lymphocytes

3.3.2. Cytotoxicity of 16 (2,4-Dinitrophenylhydrazine) Isosteviol

As evaluated from the half maximal inhibitory concentration IC50, 16 (2,4-dinitrophenyl

hydrazine) isosteviol showed IC50 value of 781.2 μM (95 % CI= 240.8 to 2535 μM). This is

slight toxicity to human peripheral blood lymphocytes (PBLs) after 48 hours exposure. Figure

3.3 shows the concentration dependant inhibitory effects of the compound on PBLs. The

cytotoxicity of 16 (2,4-dinitrophenyl hydrazine) isosteviol with IC50 value of 781.2 μM (95 %

CI= 240.8 to 2535 μM) was considerably less than that of the standard drug miltefosine 272.2

μM (95 % CI= 143.6 to 515.7 μM). Concluding the cytotoxicity estimation against human

peripheral blood lymphocyte cells, it was revealed that, in both cases, these test compounds

along with miltefosine showed minimum tolerable cytotoxicity.

Page 84: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

62

Figure 3.2. Cytotoxicity of 16 (2,4-dinitrophenylhydrazine) isosteviol

3.3.3. Cytotoxicity of 17 Hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol

This structurally different Isosteviol derivative containing a hydroxyl group, was found more

cytotoxic than the 16 (2,4-dinitrophenylhydrazine) Isosteviol, its parental skeleton. The IC50

value of this compound was 294.1 μM (95 % CI= 177.4 to 487.5 μM) as compared to that of the

miltefosine 272.2 μM (95 % CI= 143.6 to 515.7 μM) i.e., 9.2% less cytotoxic than miltefosine.

Figure 3.4 displays the concentration dependent percent inhibition of PBLs in a 48 hours

exposure time. The inhibitory effects of the compound increased with concentration.

Page 85: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

63

Figure 3.3. Cytotoxicity of 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol against

human peripheral blood lymphocytes.

3.3.4. Cytotoxicity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol

The compound showed limited toxicity with an IC50 value of 421.8 μM (95% CI= 211.3 to

842.1 μM) i.e., 35.46% less cytotoxic than that of the miltefosine. Figure 3.5 shows the impact of

this compound on the growth of the cultured PBLs in vitro. The fifty percent inhibitory

Page 86: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

64

concentration of this compound certifies adequate, if not complete, safety of the compound.

Figure 3.4. Cytotoxicity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol against

human peripheral blood lymphocytes.

3.3.5. Cytotoxicity of Actinomycin D

Actinomycin D (C62H86N12O16) is a recognized antibiotic of the actinomycin group that

demonstrates high antitumor and antibacterial action. This has been extensively used in medical

practice since 1954 as an anticancer medication for handling numerous tumour types and it is

also a valuable contrivance in cell biology and biochemistry. It attaches to DNA and obstructs

RNA production (transcription), so protein synthesis also drops.

As revealed by the IC50 value 195.8 μM (95% CI=135.3 to 283.2 μM), the Actinomycin D

showed greater cytotoxic effects as compared to miltefosine and the Isosteviol derivatives. The

cytotoxicity of the Actinomycin D showed a concentration dependant increase as is revealed in

the figure 3.5. Actinomycin D being more cytotoxic than the standard drug, has inhibitory effects

Page 87: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

65

which are still not that much perfect to the host cells. However, the compound can be considered

harmless antileishmanial contender at lower doses.

Figure 3.5. Cytotoxicity of Actinomycin D against human peripheral blood lymphocytes.

3.3.6. Cytotoxicity of Actinomycin Z3

Actinomycin Z3 (C62H83ClN12O18) compound gave an IC50 value of 210.1 μM (95% CI= 145.2

to 304.1 μM) which is comparable to actinomycin D and less than miltefosine (Figure 3.6). As

such the results of this study showed safety of the compound for human PBLs. The fifty percent

inhibitory concentration of cytotoxicity (IC50 value of 210.1 μM) is such that the compound can

be a harmless candidate for its antileishmanial action as well as it may be used for other

antiparasitic activities also. Inhibitory effects of this compound are negligible in the living

system of the host as shown by its effect on PBLs.

Page 88: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

66

Figure 3.6. Cytotoxicity of Actinomycin Z3 A against human peripheral blood lymphocytes

3.3.7. Cytotoxicity of Actinomycin Z5

Actinomycin Z5 gave an IC50 value of 234.9 μM (95% CI= 155.5 to 354.9 μM). The compound

is less cytotoxic than miltefosine and Z3. So can be safe for the host cells in vitro. As is revealed

by figure 3.7, the compound gives less than 50% cytotoxicity even at the highest concentration

used at 48 hours incubation.

Page 89: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

67

Figure 3.7. Cytotoxicity of Actinomycin Z5 against human peripheral blood lymphocytes.

3.4. Discussion

Nature is an outstanding unit for novel therapeutic challengers, as marvelous chemical diversity

is found in thousands of species of microorganisms, animals, plants and marine organisms. Plant-

derived metabolites and phytochemicals second the microbial metabolites as candidate

therapeutic agents. Although it has been at the advent of 20th century that microbial agents

surpassed the plant extract-based cures and remedies that have a history going back to ancient

Mesopotamian and Egyptian civilizations about 2600 and 2900 BC respectively. There are

records of hundreds of drugs, primarily of plant origin, which have been used by Mesopotamian

and Egyptian societies (Atanaso et al., 2015). Bacterial and fungal secondary metabolites

emerged at the discovery of penicillin and were amongst the major suppliers of the marketed

medications. Most of the available anti-infectious chemotherapeutics are either bacterial or

fungal metabolites. These metabolites, apart from having antibacterial, antifungal, antiviral,

Page 90: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

68

anticancer and anti-enzyme potentials, have also been shown to possess a variety of different

bioactivities including their uses as herbicides and source of biodegradable plastics (Duke et al.,

2000; Bérdy, 2005; Mooney, 2009).

Actinomycins are a group of 35-50 secondary metabolite compounds of actinomycetes bacteria

especially Streptomyces spp. Members of the Actinomycin group possess antibiotic, cytostatic,

anti-neoplastic and hormone-like stimulatory effects. Their antimicrobial effects are mediated by

blocking DNA synthesis when they intercalate to the minor grove (Grand et al., 1972; Hollstein,

1974; Cooper and Braverman, 1977; Zotchev, 2012) and prevent DNA mediated RNA synthesis

which is believed to be the possible mechanism behind their anti-neoplastic nature (Quigley et

al., 1980; Nunn et al., 1991).

Actinomycins, regarding their biological activities, are second to none but their cytotoxic effects

are the main hinderance in their path to the list of approved therapeutics (Kamitori and

Takusagawa, 1994). However, being harmful to the DNA of various cell types, they are still in

contention for antimicrobial and anti-cancer research. Restructuring through chemical

modification of the peptide or chromophore components has been found to improve therapeutic

nature of these compounds (Sengupta et al., 1985; Anthony and Helmut, 2011). Actinomycin D

have been found to show higher toxicities to renal, cortical and medullary cells, myocardial cells,

pulmonary histocytes and spleen cells in vitro. When used in combination with human

recombinant TNF-α against various cell types, renal medullary cells showed high sensitivity as

compared to liver cells, cardiac muscle cells and bladder cells (Aoki et al., 1998). Actinomycin

C have been found to cause morphological alterations in lymphocytes at a concentration of 5-

10μg/mL. It has been known to cause obstruction of RNA synthesis in lymphocytes at doses as

low as 1μg/mL (Lundgren and Mӧller, 1969). Actinomycin D has been found to cause inhibition

of bovine oocytes maturation by causing chromosomal modifications at a concentration of 2.5-

5μg/mL after 14-hour incubation. Lower concentration, however, caused little chromosomal

alterations and maturation impairment at the same exposure period (Moura et al., 2008).

Similarly, in mice model at lower dosages, actinomycin D caused reduced suppression of

transcription e.g., 0.25mg/kg body weight, five percent genes were found supressed (Siboni et

al., 2015). The present study reveals more or less comparable findings regarding cytotoxic

Page 91: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

69

effects of actinomycins on normal human peripheral blood lymphocytes. These microbial

metabolites deliver both therapeutic and toxic effects at minor concentrations.

The glycoside metabolites of Stevia rebaudiana (also called as sweet leaf is flowering plant in

the family Asteraceae) are one of the major non-caloric sweetening food additives used around

the world. Among these, steviol, stvioside, rebaudioside and Isosteviol are notable metabolites.

Presently these and their natural or artificial derivatives are under trials to assess their

bioactivities beyond sweetening. Regarding their adversities to human health, variable opinions

and evidences exist. They have, however, been characterized in various in vitro and in vivo

studies with no teratogenicity, cytotoxicity, mutagenicity or carcinogenicity (Takasaki et al.,

2009; Momtazi et al., 2016) or behavioural, haematologic, clinical and histopathological effects

(Zhang et al., 2017). Steviosides has been demonstrated to cause no cytotoxicity in peripheral

blood mononuclear cells (Noosud et al., 2017). In the present study for all the three hydrazine

derivatives of Isosteviol the inhibitory effects on lymphocytes were found to be in much higher

micromolar concentrations. The 50% inhibitory concentrations were found to be more than 400

μM giving indication of their nonaggressive nature to human PBLs and tissues. Steviol and

isosteviol derivatives of alkyl amide dimerization have been revealed to possess in vitro anti-

neoplastic potency against different cell lines. One study, however, found these compounds to be

less toxic to normal human embryonic lung cells (Lin et al., 2004). Animal studies have found

crude extracts and polyphenols from stevia to alleviate liver and kidney damage in streptozotocin

induced diabetes (Shivanna et al., 2013). Although hundred percent literature available regarding

health safety of the natural Stevia metabolites and synthetic analogues is not stamping a safe

nature of these compounds, most of the studies have found no or negligible adversities to normal

cells and tissues. Synthetic analogues, however, have been found to be comparatively more

aggressive along with higher therapeutic capabilities against cancer and infectious agents.

3.4. Conclusion

The present study found actinomycins to be more toxic to human peripheral blood lymphocytes

and some with comparable cytotoxicity to standard drug Miltefosine. For all the three candidates

of this group, the fifty percent inhibitory concentrations were less than 1μg/mL in in vitro setup

to human peripheral blood lymphocytes.

Page 92: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

70

Hydrazine derivatives of Isosteviol were less cytotoxic to human peripheral blood lymphocytes

in vitro. Derivatives of isosteviol are biologically energetic and are one of nature exceptional

compounds with hydrophobic and hydrophilic possessions. Their skeletal structure can easily

be used in the synthesis of a number of other useful derivative compounds and to design

active, selective and polyfunctional Isosteviol analogues for the therapy of leishmaniasis and

other infections.

Page 93: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

71

Chapter 4. Antileishmanial Compounds Screening in vitro

Cutaneous leishmaniasis has assumed an epidemic status in our province Khyber Pakhtunkhwa

in Pakistan and according to WHO local office and ministry of health about 23,760 patients are

registered in January-February 2019 only. An international medical charity organization

“Medicines Sans Frontieres” (MSF) has started treating cutaneous leishmaniasis patients with

meglumine antimonate but have several side effects (Figure 4.1). There is a dire need to stem the

tide of fast growing cutaneous leishmaniasis in Pakistan. This investigation is an effort to stem

the tide of growing cutaneous leishmaniasis and to balance toxicity with efficacy, as a

predictable dosing plan calls for intermittent application of an antileishmanial compound at or

near the maximum tolerable dose.

Figure 4.1. Cutaneous leishmaniasis patients are treated in Peshawar Hospital with intralesional

injection of meglumine antimonate (https://www.dawn.com/news/1461261).

In vitro screening is an important mean of preliminary evaluation of biological and antiparasitic

or antimicrobial activity of compounds. Such lab dish evaluations can give valuable insights to

the in vivo drug interactions with the living systems (Volpe et al., 2014). Biotechnological

approaches are desirable to improve superior medications against leishmaniasis. Though

paromomycin and miltefosine were registered as therapeutic agents against Leishmania donovani

infection in the previous decade, the leishmanicidal medication arsenal still necessitates

upgrading, mainly in the zone of oral antileishmanials for cutaneous, mucocutaneous and

visceral leishmaniasis. Numerous novel preparations and compounds have demonstrated

Page 94: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

72

encouraging efficiency in animal models infected with various Leishmania species. In this

investigation, we tested some existing drugs used for the treatment of other diseases as new

antileishmanials against Leishmania tropica promastigotes and amastigotes in vitro. In the

present study, antileishmanial effects of six compounds and standard drug miltefosine were

analysed against both the promastigotes and axenically transformed amastigotes of Leishmania

tropica in vitro.

4.1. Materials and Methods

4.1.1. Culture of the Leishmania tropica KWH23 Promastigotes

Promastigotes of Leishmania tropica KWH23 were grown aseptically in RPMI 1640 growth

medium supplemented with 10% heat-inactivated foetal bovine serum (HI-FBS) and 100 μg/mL

streptomycin and 100 IU/mL penicillin at a density of 1×105/mL at 26°C under anaerobic

conditions. The Sigma-Aldrich RPMI 1640 was already added with L-glutamine an unstable

necessary amino acid needed at a final concentration of 29.2 mg/mL. For setting up a

Leishmania tropica KWH23 growth curve, a culture was initiated between 1x105 to 1x106

parasites/mL and Leishmania cells were counted daily for 7 days. The growth was monitored for

a week until the culture reached the stationary phase of growth. When mentioning to the phase of

growth in Leishmania tropica KWH23 cultures, stationary stage was obtained at ~2-3 x 107

promastigotes/mL. It was these stationary promastigotes that were subsequently utilized for anti-

promastigote assays and for generation of the axenic amastigotes.

4.1.2. Generation of Axenic Amastigotes

Stationary culture of Leishmania tropica KWH23 was harvested by centrifugation and diluted to

1×106/mL in RPMI 1640 medium fortified with 20% HI-FBS and 1% v/v streptomycin and

penicillin antibiotics. The pH of the RPMI 1640 medium was adjusted to 4.4 using concentrated

phosphoric acid as recommended in Sigma-Aldrich cell culture manual. At the acidic pH the

medium colour turned light yellow. The Leishmania tropica KWH23 promastigotes, during

transformation, were maintained at 37°C in humidified conditions in the absence of CO2. The

transformation process was monitored through Giemesa staining daily. Stationary promastigotes

after 10 days in culture without medium changes (as mentioned previously in section 2.1.4) was

Page 95: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

73

converted to fully round to oval aflagellated cells (axenic amastigotes) with more than 90%

viability were utilized for sensitivity assays.

4.1.3. Antileishmanial Activity

Both promastigote and amastigote stages of Leishmania tropica KWH23, in separate

experiments, were seeded at 1×106/well in 96-well flat bottom plates in 200 μL of RPMI1640

medium fortified as mentioned above for both the stages of the parasite. The test compounds and

the standard drug miltefosine were added in 8 different concentrations i.e., 0.02, 0.2, 2, 20, 25,

50,75 and 100 μM respectively. All the experiments were performed in triplicate. A negative

control was run with RPMI 1640 growth medium with 1% DMSO, the solvent used in the test

compounds. Both promastigotes and amastigotes were exposed to the compounds for 48 hours

under the respective conditions of temperature and pH for both the stages. After 48 hours

incubation, 20 μL of XTT solution was added per well and plates were covered with alluminium

foil and incubated for 2 hours under the respective conditions of temperature (26-37ᴼC) for both

promastigotes and amastigotes. After 2 hours, the absorbance in terms of viability was read in

Bio-Tek ELx800 ELISA microplate reader through GEN5 software at 490 and 630 nm

wavelength. The formazan assembly by XTT reduction is soluble in water and therefore was

directly detected in promastigotes and amastigotes supernatants so was straightforwardly

calculated by colorimetric examination.

4.1.4. Data Analysis

The absorbance statistics was first manipulated by deduction of the 630nm value from 490 nm

value for each well. The values obtained after subtraction were then processed in Microsoft

Excel 2016 and converted to percent inhibition by using the standard formula (control-

treated/control×100). The percent inhibition values for each compound against promastigote and

amastigote stages of Leishmania tropica KWH23 were then processed by GraphPad Prism 6

software to calculate the IC50 values for the test compounds and the standard drug miltefosine.

Page 96: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

74

4.2. Results

4.2.1. Antileishmanial Activity of Miltefosine

Miltefosine, the only oral approved therapy for visceral and cutaneous leishmaniasis, was used as

standard positive control in the present study. Miltefosine (C21H46NO4P) is a phospholipid with

elimination half-life of 6 to 8 days and Anatomical Therapeutic Chemical (ATC) classification

number L01XX09 as maintained by WHO. It gave IC50 values of 10.80μM (95% CI=9.114 to

12.81 μM) and 1.245 µM (95% CI=0.7250 to 2.138 μM) against promastigotes and amastigotes

respectively. Figure 4.2 shows the leishmanicidal activity of the drug against both the

promastigotes and amastigotes. As is revealed by the figure below, its killing ability increased

with concentration against both the stages of the parasite. However, greater degree of variability

in the killing of both the forms of the parasite was seen from 0.02-20 μM than from 25-100 μM.

It caused maximum killing of both promastigotes and amastigotes at the highest concentration

over 48-hour exposure. The present investigation reveals miltefosine as potent therapeutic

against Leishmania tropica. The amastigotes, as given by the IC50 values and the figure below,

were found more susceptible to the drug as compared to the promastigotes.

Figure 4.2. Antileishmanial activity of miltefosine against promastigotes (A) and amastigotes (B)

of L. tropica KWH23.

Page 97: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

75

4.2.2. Antileishmanial Activity of 16 (2,4-dinitrophenylhydrazine) Isosteviol

Figure 4.3 shows the antileishmanial activity of 16 (2,4-dinitrophenylhydrazine) isosteviol. The

IC50 values of the compound against both the promastigotes and amastigotes were 7.098μM

(95% CI=5.328 to 9.455μM) and 4.447μM (95% CI= 2.788 to 7.094μM) respectively.

Leishmanicidal activity of the compound increased with the concentration showing greater

variability when moving from 0.02-20μM to 25-100μM. The compound gave higher activity

against the amastigotes than the promastigotes. As revealed by the fifty percent inhibitory

concentrations (IC50) of the compound 16 (2,4-dinitrophenylhydrazine) isosteviol, it is

comparable with those to miltefosine so it can be a good antileishmanial candidate.

Figure 4.3. Leishmanicidal activity of 16 (2,4-dinitrophenylhydrazine) Isosteviol against

promastigotes (A) and amastigotes (B) of L. tropica KWH23.

4.2.3. Antileishmanial Activity of 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol

As revealed in Figure 4.4, the IC50 values of 5.603μM (95% CI= 4.628 to 6.784μM) and

5.033μM (95% CI= 3.189 to 7.945μM) against promastigotes and amastigotes of L. tropica

KWH23 were detected by the compound 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol.

Page 98: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

76

This clearly depicts that 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol possesses

virtuous antileishmanial bustle in vitro as compared to the standard antileishmanial

alkylphospholipid miltefosine. From the present results, it can be concluded that 17 hydroxy, 16

(2,4-dinitrophenylhydrazine) Isosteviol may be proved as an ideal antileishmanial agent

particularly in cutaneous leishmaniasis caused by L. tropica and this may be a central compound

for the development of potent antileishmanial in future. Consequently, development of innovative,

active, and harmless antileishmanial compounds, with fewer side effects, is a main priority of this

health investigation.

Figure 4.4. Antileishmanial activity of 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol

against promastigotes (A) and amastigotes (B) of L. tropica KWH23.

4.2.4. Antileishmanial Activity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol

The antileishmanial activity of the compound benzyl ester 16 (2,4-dinitrophenylhydrazine)

Isosteviol is revealed in figure 4.5. A concentration dependant response of both the

promastigotes and amastigotes of L. tropica KWH23 is clear from the figure 4.5. As revealed in

the figure 4.5, IC50 values of benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol are

10.71μM (95% CI= 8.611 to 13.31μM) and 6.794μM (95% CI= 4.248 to 10.87μM) against the

Page 99: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

77

promastigotes and amastigotes of L. tropica KWH23 respectively. Like previously mentioned

compounds used in this investigation benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol is

more active against the amastigote forms as compared to metacyclic promastigotes. The

antipromastigote activity of the compound is almost similar to that of the miltefosine, however,

regarding its antiamastigote activity benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol is

less effective than the miltefosine. The overall results, however, reveal that the compound may

be a potent candidate for the antileishmanial drug development.

Figure 4.5. Antileishmanial Activity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol

against promastigotes (A) and amastigotes (B) of L. tropica KWH23.

4.2.5. Antileishmanial Activity of Actinomycin D

Figure 4.6 represents the leishmanicidal activity of Actinomycin D (M1) against both the

promastigotes and amastigotes of L. tropica KWH23. Actinomycin D has been expansively

exploited in medical exercise since 1954 as an anticancer medicine for management of abundant

cancer types and it is correspondingly a valuable gadget in cell biology and biological chemistry.

It hinders RNA synthesis (transcription), so protein synthesis also drops. As indicated by the IC50

values 8.739 μM (95% CI= 6.675 to 11.44 μM) and 2.135 μM (95% CI= 1.419 to 3.211μM) for

Page 100: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

78

promastigotes and amastigotes forms, the compound possesses good antileishmanial activity.

The leishmanicidal effects against both the promastigotes and amastigotes are parallel with that

of the standard drug miltefosine. The higher inhibitory activity against amastigote forms of L.

tropica KWH23 reveals that the compound may probably become a good antileishmanial

candidate after extensive in vitro and in vivo investigations in future.

Figure 4.6. Antileishmanial activity of Actinomycin D (M1) against promastigotes (A) and

amastigotes (B) of L. tropica KWH23.

4.2.6. Antileishmanial Activity of Actinomycin Z3

Figure 4.7 depicts the antileishmanial activity of Actinomycin Z3, also designated as M2. The

compound gave promising antileishmanial activity against metacyclic promastigotes as well as

amastigote stages of the L. tropica KWH23 parasite. Against both the stages of L. tropica,

Actinomycin Z3 gave efficacious results than the miltefosine with IC50 values of 5.5μM (95%

CI= 3.811 to 7.939μM) and 1.760μM (1.136 to 2.728μM) respectively. Activity against

amastigotes was, however, higher as compared to the metacyclic promastigotes. With the current

in vitro results, the compound Actinomycin Z3 is proposed for further antileishmanial activities

and it is expected to come up as a potential leishmanicidal agent.

Page 101: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

79

Figure 4.7. Antileishmanial activity of Actinomycin Z3 (M2) against promastigotes (A) and

amastigotes (B) of L. tropica KWH23.

4.2.7. Antileishmanial Activity of Actinomycin Z5

The Actinomycin Z5 (M3) gave the IC50 values of 9.529 μM (95% CI= 7.354 to 12.35μM) and

1.691μM (95% CI= 0.9559 to 2.991μM) against the promastigotes and amastigotes of L. tropica

KWH23 respectively (Figure 4.8). The compound showed less cytotoxicity than miltefosine and

Z3 as tested against human peripheral blood lymphocytes. This Actinomycin Z5 may be

harmless for the host cells as shown by in vitro results as revealed in figure 3.8. The compound

showed greater activity against amastigotes than promastigotes as is clear from the mentioned

fifty percent inhibitory concentrations represented in figure 4.8. The overall IC50 activity of the

compound is comparable to that of the miltefosine. The present in vitro results elucidate that

actinomycin Z5 compound may be an ideal candidate against both the life cycle stages of the

other species of Leishmania genus prevalent in Pakistan like L. donovani, L. infantum and L.

major too.

Page 102: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

80

Figure 4.8. Antileishmanial activity of Actinomycin Z5 (M3) against the promastigotes (A) and

amastigotes (B) of L. tropica KWH23.

4.3. Discussion

Although search for new therapeutics against leishmaniasis has been there since the discovery of

antimonials and still lingers, but no novel and effective agent has been discovered with limited

side effects. First oral antileishmanial drugs like miltefosine and paromomycin were registered as

clinical compounds against visceral leishmaniasis in the preceding decade, the antileishmanial

medication battery still needs upgrading, predominantly in the area of oral antileishmanial

medications for both cutaneous and visceral form of the disease.

In this hunt for new antileishmanials, phytoproducts and antimicrobial metabolites are on the

uppermost priority among the biological contenders. Traditional ethnobotanic practices propel

inflight to the mission for exploring plant products to skim out new antimicrobials together with

antileishmanials (Iwu et al., 1994).

Page 103: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

81

4.3.1. Mechanism of Action of Miltefosine

Miltefosine is a main therapeutic development as it is the only oral drug for the treatment of

visceral form of leishmaniasis caused mainly by L. donovani in India and Pakistan. Miltefosine

development displayed that public-private establishment is a practicable model for encouraging

exploration and development in neglected tropical diseases like leishmaniasis (Sunyoto et al.,

2018). Miltefosine obstructs the synthesis of phosphatidylcholine and affects the Leishmania

mitochondrion by cytochrome-c oxidase inactivation. It has been testified that the interruption of

the intra-leishmanial Ca2+ homeostasis characterises a significant entity for the action of

antileishmanial drugs. Pinto-Martinez et al. (2018) have demarcated a plasma membrane

Ca2+ channel in Leishmania mexicana, which is similar to voltage-gated Ca2+channel present in

human cells. This Leishmania Ca2+ channel is activated by miltefosine, particularly in L.

donovani in vitro. Miltefosine strongly affect the acidocalcisomes from L. donovani, making

quick alkalinisation of these organelles. Acidocalcisomes are acidic organelles which resemble

lysosome-related organelles of mammalian cells and contains high concentration of phosphorus

amalgamated with calcium and accompanying cations. Acidocalcisome contribute in calcium

homeostasis, management of intracellular pH and osmoregulation. Acidocalcisomes resemble

lysosome having comparable size distribution, acidic possessions, and together having

phosphorus and calcium. So miltefosine interaction and endocytosis to Leishmania tropica in

present study may have all the above-mentioned mechanism responsible for the demise of

Leishmania tropica promastigotes and amastigotes by quick alkalinisation of acidocalcisomes

and thereby disrupting calcium homeostasis, management of intracellular pH and

osmoregulation.

Page 104: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

82

Figure 4.9. Leishmania promastigotes (A) and amastigotes (B) showing acidocalcisomes

(https://www.sciencedirect.com/science/article/pii/S0020751907001038#fig1).

4.3.2. Antileishmanial Action of Isosteviol and Actinomycin D

A variety of different diterpenoids and their derivatives have been tested for their biological

activities (Kubo et al., 2004; Zhu et al., 2013; Wang et al., 2018). All the three Isosteviol (an

ent-beyrene diterpenoid) derivative used in the present study showed good activity against both

the flagellated promastigotes and amastigote forms of the Leishmania tropica KWH23. Natural

non-caloric glycosides from Stevia rebaudiana and many other plants which includes stevioside,

steviol and Isosteviol and their derivatives have been variously tested for their anticancer,

antibacterial, antifungal, antiprotozoal and antioxidant potential (Malki et al., 2014). Isosteviol

and its derivatives have been proposed to possess broad spectrum biological activities.

Antituberculotic potential of Isosteviol, steviol and their derivatives have been confirmed by

several in vitro investigations. The minimal inhibitory concentrations (MIC) found in these

studies have been mostly in the range of 0.3-10 μM for different derivatives. Some of these,

Page 105: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

83

however, showed noticeably lower activity (Kataev et al., 2011; Khybullin et al., 2012;

Garifullin et al., 2016).

Whole extracts of Stevia rebaudiana has been found to effectively inhibit the growth of plague

spirochete Borrelia burgdorferi in vitro. These extracts were also found significantly effective

against biofilms of the microbes which normally contain more resistant cells (Theophilus et al.,

2015). Isosteviol from other plants like Pittosporrum tetraspermum has also been found much

effective against different pathogens including Staphylococcus species, Escherichia coli,

Salmonella typhi, Klebsiella pneumoniae and Pseudomonas aeruginosa. It has shown different

degrees of antifungal activities against Candida albicans, Aspergillus niger and Trichophyton

mentagrophytes (Al-Dhabi et al., 2015). Similarly, ent-kaurane diterpenoids, close relatives of

the Isosteviol, have shown antimicrobial activity against Gram-positive bacteria including

Bacillus subtilis, Streptococcus mutans, Streptococcus aureus, Brevibacterium ammoniagenes,

and Propionibacterium acnes. They, however, not been found effective in inhibiting Gram-

negative bacteria and fungi (Kubo et al., 2004). When tested against hepatits B virus, Isosteviol

inhibited the secretion of both HBsAg and HBeAg giving EC50 values of 4.8 and 5.9μg/mL

respectively. At concentration greater than 5 μg/mL, it reduced the synthesis of viral DNA

(Huang et al., 2014). Another derivative of Isosteviol IN-4 [N-(propylcarbonyl)-4aamino-19-

nor-ent-16-ketobeyeran], effectively inhibited the secretion HBsAg and HBeAg as well as the

DNA production at a concentration as low as 5.67 μg/mL (Huang et al., 2015).

Diterpenoids and terpenes have also been revealed for their antiprotozoal activity against the

major protozoan pathogens including Plasmodium, Trypanosoma, and Leishmania species and

other neglected tropical diseases (de Alencar et al., 2017). These studies have mostly been done

in vitro. Candidates from ent-kaurane diterpenoids, close relatives of Isosteviol, have shown

activity against Plasmodium parasites. Some of the compounds have been of substantial

importance regarding their antiprotozoal effects in micromolar concentrations ranging from 4.5-

10.4μM against chloroquine resistant W2 strain of Plasmodium. It has been noted that the

substitution of carboxylic group enhances the antiplasmodial effects. Methyl kaurenoate and

methyl iso-kaurenoate have been revealed to show higher activity against Plasmodium

falciparum W2 (IC50 8.2 and 8.8 μM respectively) than the kaurenoic acid and grandiflorenic

Page 106: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

84

acid (IC50 21.1 and 23.3 μM respectively) (Batista et al., 2013). This kind of activity of ent-

kaurane diterpenoids have also been noted against Trypanosoma cruzi (Batista et al., 2007).

Actinomycins have been extensively investigated for their biological potencies including

antimicrobial and antineoplastic activities. Actinomycin D is an approved therapy for several

different types of malignancies. In an in vitro study, actinomycin Z3 showed highest activity

against Streptococcus aureus at a concentration of 50 μg/mL with an inhibition zone of 10 mm

followed by actinomycin Z5 (8 mm), actinomycin Z1 (7 mm) and actinomycin Z6 (0 mm). None

of these compounds, however, showed inhibition of Candida albicans (Dong et al., 2018). As

evaluated by cross-streak assay, actinomycin D caused inhibition of different species of fungi

and Streptomyces mutabilis with inhibition zone from 19-45 mm. Actinomycin D was also found

effective as biocontrol agent against chocolate spot (Botrytis cinerea) and Fusarium wilts and

rots causing Fusarium oxysporum (Toumatia et al., 2015).

Streptomyces metabolites (actinomycin Z5) possesses wide antimicrobial activities including

several important protozoan parasites including Plasmodium, Leishmania, Trypanosoma and

Entamoeba species. When tested against promastigotes of three different species of Leishmania

i.e., L. tropica, L. enrietti, and L. donovani, actinomycin D was found to cause inhibition on

translation of heat shock proteins at 40 μg/mL (Lawrence and Robert-Gero, 1985). The

inhibitory concentrations reported in the present study reflect close similarity to the mentioned

study. Along with protein inhibition, it also found to cause fragmentation of DNA in Leishmania

amazonensis promastigotes (Mendes et al., 2016). Inhibition of translation by preventing

transcription of genes crucial for pre-mRNA maturation of mRNA have been recently revealed

in Plasmodium falciparum. Its major targets were falcipain-2, FP-21-cys peroxiredoxin and

Plasmodium falciparum cleavage and polyadenylation specificity factor subunit 3 (pfCPSF). The

inhibition has been described at 80 and 320 μg/mL for the two chloroquine resistant strains W2

and Dd2 of Plasmodium falciparum (Sonoiki et al., 2017). Actinomycin D based inhibition of

protein and DNA synthesis have been confirmed in other study at picomolar (50 pM)

concentration (Diekmann-Schupert and Franklin, 1990). It also has been proposed to cause

transcription inhibition and mitochondrial dysfunction in P. falciparum (Pérez-Moreno et al.,

2016). Actinomycin D could cause 58% and 53% reduction in Plasmodium vinckei parasitaemia

at 10 μg/kg body weight in mice respectively once and twice a day (Fink and Goldberg, 1969). It

Page 107: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

85

also caused growth inhibition and morphological changes in Trypanosoma cruzi at 10, 20 and 50

μg/mL for 24-hour treatment in vitro. The parasite however, remained alive for at least 2 weeks

after this treatment (Zaverucha et al., 2003). The parasites treated with actinomycin D at

50μg/mL did not caused immunosuppression of both cellular and humoral immune response in

mice suggesting its potential use as a vaccine candidate (da Cruz et al., 1989). Actinomycin Z

has been found to effectively eradicate Entamoeba histolytica at 100 μg/mL in vitro. In almost

all the mentioned previous investigations, these compounds have been found effective in

inhibiting the various protozoans at low micromolar concentrations showing much similar results

to the present work. Further in vitro as well as in vivo studies are, however, needed to find the

mechanism of action and toxicity of the actinomycins using different cell types and animal

models.

Page 108: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

86

Chapter 5. Evaluation of the Compounds Genotoxicity against Human

Lymphocytes

5.1. Introduction

Pakistan is rich in traditional remedial flora with an enormous biodiversity which offers a good

occasion to explore the plants to hunt new, effective, less toxic and harmless therapeutic agents.

One of the several challenges faced by the biomedical researchers is the quantification of

physical damage to DNA by many physical (Osipove et al., 2004) and chemical agents. These

agents include food flavours, pharmaceuticals, cosmetics and radiations, to which living system

is either directly or indirectly exposed. In this regard, Comet assay is one of the simple and

convenient technique to assess DNA damage by electrophoresing whole nuclei on microscope

slides from cells, either experimentally exposed to an agent in vitro or isolated from living body

naturally exposed to the agent (Ostling and Johnson, 1984; Reus et al., 2012). The term "comet"

denotes to the design of DNA movement through the electrophoresis gel, that commonly

resemble a comet (bright object with long tail). The importance of Comet Assay can be judged

from the fact that every year International Comet Assay Workshop is held at different countries.

This year 13th International Comet Assay Workshop (ICAW 2019) will be held in Pushchino, a

town in Moscow Oblast in Russia from 24-28 June 2019 (www.mutagenesisambiental.com/13th-

international-comet-assay-workshop/). These workshops are a succession of scientific meetings

dealing with practical and theoretical aspects of the Comet Assay. The original publication

relating such an assay was published 35 years before in 1984 by Swedish investigators O.

Ostling and K. J. Johanson in Biochemical and Biophysical Research Communications. This

assay was further established by Singh et al. (1988) and afterward this assay has been

extensively used in measuring DNA damage. The present study is one such attempt to assess

DNA damage caused by Actinomycin (actinomycetes metabolite) and Isosteviol derivatives

(plant metabolites) in human peripheral blood lymphocytes (PBLs).

5.2. Methodology

Genotoxic effects of the Actinomycin and Isosteviol derivatives on human peripheral blood

lymphocytes were evaluated using alkaline comet assay protocol originally developed by Ostling

and Johnson, 1984, modified by Singh et al. (1988) and further improved by Osipov et al. (2014)

Page 109: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

87

and Khisroon et al. (2015). In this technique the cells are first entrenched in agarose on a clean

glass slide and are then lysed by using detergent to nucleoids having supercoiled coils of DNA

connected to the nuclear milieu. Then electrophoresis at raised pH results in structures like

comets, as observed under fluorescence microscope; the strength of the comet tail comparative to

the head reflects the quantity of DNA disruptions. A variety of different alternatives including

reverse mutation assay, chromosomal aberration test and micronucleus tests, all with efficiencies

and limitations, are used for genotoxicity evaluation. Comet Assay is, however, easy and

comparable to the rest.

5.2.1. Application of Test Compounds to Lymphocytes for Genotoxicity

To check the genotoxicity of the test compounds, lymphocytes were incubated for 48 hours with

Actinomycin D, Actinomycin Z3, Actinomycin Z5, 16 (2,4-dinitrophenylhydrazine) Isosteviol,

17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol, Benzyl ester 16 (2,4-

dinitrophenylhydrazine) Isosteviol and miltefosine (standard positive control). The

concentrations used were100μM, 25μM and concentration caused 50% inhibition (IC50) in

axenic amastigotes. The IC50 concentrations were 2.135, 1.76, 1.691, 4.447, 5.033, 6.794 and

1.245μM respectively for Actinomycin D, Actinomycin Z3, Actinomycin Z5, 16 (2,4-

dinitrophenylhydrazine) Isosteviol, 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol,

Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol and miltefosine. Only cultured human

peripheral blood lymphocytes and 1% DMSO were used as negative control and were denoted as

“Plain”. Three different experiments were done in triplicate to validate the results.

5.2.2. Slide Preparation and Mounting of Treated Lymphocytes

After required exposure to the test compounds, a 40μL of cell suspension (1x105/mL) was mixed

with 130μL of molten low melting point agarose (LMPA) in a 3 mL sterile Eppendorf tube. This

mixture was then smeared on a normal melting agarose (NMA) pre-coated slide. Pre-coating was

done 15 hours before by simply plunging a degreased slide in molten 0.7% NMA. Two smears

were prepared per slide. Each smear was overlaid by a separate 22x22 mm clean sterile coverslip

avoiding bubbles. The smear was solidified on an ice pack for 5 minutes and the coverslip was

removed by careful side pushing with a finger.

Page 110: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

88

5.2.3. Lysing of Lymphocyte Cells

Lysis of the human peripheral blood lymphocytes was performed in a working lysing solution

prepared freshly from stock lysing solution (2.5 M NaCl, 100 mM Na2-EDTA, 10 mM Tris,

adjusted to pH 10) by adding 1% Triton X-100 and 10 % DMSO. Lysing was performed for 2-24

hours at 4°C and cellular protein was isolated.

5.2.4. Single Cell (SC) Gel Electrophoresis

Single cell gel electrophoresis was performed in buffer prepared by mixing 300 mM sodium

hydroxide and 1 mM EDTA. The pH of the buffer was then adjusted to 13. To unwind the DNA

and to expose alkali labile regions, slide was kept immersed in buffer for 20 minutes without

switching the electrophoresis unit on. After unwinding, the electrophoresis was performed for 30

minutes at 4°C on 300mA currents at 25 V.

5.2.5. Neutralization and Fixation

Three washes of 5 minutes each were done with neutralization buffer (400 mM Tris, pH 7.5) and

slides were then fixed in 99.5% ethanol for 5 minutes and air dried at room temperature.

5.2.6. Staining

The slides were stained in 10% Giemsa stain for 20 minutes. Giemsa's stain is an associate of the

Romanowski cluster of stains, which was initially intended to integrate cytoplasmic (pink) staining

through nuclear (blue) staining and fixation as a lone stage for smears and thin films of blood and

other tissues. After rinsing under tap water and air drying, the comets were observed under light

microscope at low power objective (100 x magnifications), high power dry objective (400 x

magnifications) and high refractive indexed oil immersion objective (1000 x magnification

which increases the numerical aperture of the objective lens by oiling the specimen and the

objective) to record results (Opisov et al., 2014).

5.2.7. Data Interpretation and Scoring

For recording results, 100 randomly selected lymphocyte cells nuclei were counted from the dual

smear for each slide. The cells were then assorted to 5 different comet classes by intensity of

staining in the tail from zero (0) (no damage) to 4 (maximum/total damage). Comet scoring into

various classes was done visually. Total comet score (TCS) was determined by using formula

Page 111: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

89

TCS= 0 (n)+1(n)+2(n)+3(n)+4(n), where “n” represents number of cells in each class (Khisroon

et al., 2015).

5.2.8. Statistical Analysis

The Total comet score (TCS) values were analysed in SPSS software using two-way ANOVA

and Tuckey test for comparison of the test compound’s genotoxic potential in relation to 1%

DMSO control, plain control and standard drug i.e., Miltefosine. While at the lowest

concentration, which was the IC50 value of the compounds against axenically grown amastigotes

of Leishmania tropica as described in chapter 4, one-way ANOVA and Tuckey test were used

for comparison of genotoxicity in relation to the mentioned control and standard.

Page 112: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

90

5.3. Results

5.3.1. Genotoxicity of Miltefosine (Milt)

Miltefosine showed least genotoxicity against peripheral blood lymphocytes (PBLs). As

evaluated by comet assay, it gave total comet scores (TCS) of 10, 8 and 8 respectively at 100, 25

and 1.25μM concentrations. As revealed in figure 5.1 A, B and C, damage to only the category

of class 1 could be seen in the smears treated with all concentrations of the Miltefosine drug. As

compared to the solvent (1% DMSO) and negative (Plain) control, the genotoxicity of

Miltefosine was found to be non-significant at all concentrations used (P>0.05).

Figure 5.1. Genotoxicity of Miltefosine (Milt) at 100μM, 25μM and 1.25μM (concentration

which showed IC50 for Leishmania tropica amastigotes in Chapter 3.0).

Page 113: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

91

5.3.2. Genotoxicity of 16 (2, 4-dinitrophenylhydrazine) Isosteviol (ID)

Genotoxicity of 16 (2,4-dinitrophenylhydrazine) Isosteviol showed dependence on the

concentration of the compound used as is revealed by the total comet scores (TCS) and classes of

damage (figure 5.2). At 100μM concentration Isosteviol indicated highest damage to human

blood lymphocytes and all the 4 classes of damage were observed. A TCS value of 207.33 was

noted. As compared to the DMSO and plain control where only very little class 1 damage was

seen, the compound caused damage to class 4 level at 100μM concentration (figure 5.2 A). At 25

μM concentration, damage to the level of class 3 was detected in the treated human blood

lymphocytes cells while in DMSO and plain control damage to only class 1 was documented

(figure 5.2 B). A still higher value of 87.333 for TCS was observed.

Figure 5.2. Genotoxicity of 16 (2,4-dinitrophenylhydrazine) Isosteviol (ID) at 100μM, 25μM and

4.447μM (concentration which showed IC50 for Leishmania tropica amastigotes in Chapter 3.0).

Damage in the DMSO and plain control may be attributed to the naturally occurring apoptotic

cells in the inoculum. This little damage was persistently recorded in both types of the control.

When used at 4.447μM concentration Isosteviol (concentration which showed IC50 for

Page 114: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

92

Leishmania tropica amastigotes in Chapter 3.0), showed least genotoxic damage was observed as

revealed by a TCS value of 10.666 and damage to only class 1 level (figure 5.2-C). The

compound Isosteviol showed non-significant (P>0.05) genotoxic effects as compared to the

standard Miltefosine and 1% DMSO and negative control at all the concentration used.

5.3.3. Genotoxicity of 17 hydroxy, 16 (2, 4-dinitrophenylhydrazine) Isosteviol (ID7)

As revealed in figure 5.3, a concentration dependent variability was noted in the genotoxic

effects of 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol (ID7). A sharp waning was

seen in total comet score (TCS). It showed highest genotoxicity at 100μM concentration

successfully creating class-4 damage. Damage at the level of class 3 and 4 was, however, less

than 20% (figure 5.3 A) and TCS value of 202 was witnessed. At 25μM concentration 17

hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol showed damage to class-3 level but at the

minimum (figure 5.3 B). A TCS value of 85.333 was calculated.

Figure 5.3. Genotoxicity of ID7 at 100μM, 25μM and 5.033μM (concentration which showed

IC50 for Leishmania tropica amastigotes in Chapter 3.0).

At 5.033μM damage to the level of class 2 was created but to a minor extent while around 10%

comets were with class 1 damage (figure 5.3 C) and a TCS value of 15.333 was calculated.

Page 115: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

93

Therefore, in summary the genotoxic effects of the compound were slightest at the minimum

concentration of the 17 hydroxy, 16 (2,4-dinitrophenylhydrazine) Isosteviol. However, it was

found to possess significant genotoxicity as compared to plain at 100 and 25μM (P<0.05) but

genotoxicity became non-significant when the concentration was reduced to 5.033μM

concentration (P>0.05).

5.3.4. Genotoxicity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol (IDB5)

Genotoxicity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol (IDB5) also showed

altering scores with in consort with the concentration. Greater concentrations of the compound

had more genotoxicity in the human blood lymphocytes (figure 5.4). All the four classes of

genotoxic damage were seen at 100μM concentration while genotoxic damage to class 3 was

detected at 25μM concentration (figure 5.4 A and B). The TCS values of 166.33 and 85.33 at

100μM and 25μM concentration were acknowledged. At 6.794μM concentration Benzyl ester 16

(2,4-dinitrophenylhydrazine) Isosteviol showed a negligible damage at class 2 level with a TCS

value of 15 (figure 5.4-C). As revealed in the figure 5.4, total comet score got steep decline as

the concentration of the compound was reduced. Th compound, however, showed non-

significant level of genotoxicity as compared to the standard Miltefosine, 1% DMSO and

negative controls at all concentrations (P>0.05). Safety of the compound to cause genotoxicity is

tangled to its concentration used and causes negligible and probably restorable harm to human

blood lymphocytes at lower concentration.

Page 116: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

94

Figure 5.4. Genotoxicity of Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol at 100μM,

25μM (concentration which showed IC50 for Leishmania tropica amastigotes in Chapter 3.0).

5.3.5. Genotoxicity of Actinomycin D (M1)

Actinomycin D produced greater genotoxic effects in human blood lymphocytes in comparison

to standard drug Miltefosine and the test compound Isosteviol. Genotoxicity was, however,

greatly dependant on the concentration of the compound used. Highest value (327) of total comet

score (TCS) was detected at 100μM along with higher percentage of damage at the levels of

class 3 and 4 as depicted in figure 5.5 A. At 25μM still a higher amount of genotoxicity was

caused on the lymphocyte DNA as is revealed by the TCS of 207 and class 3 and 4 damage

(figure 5.5 B). At 2.135μM concentration, however, the genotoxic effects got much reduced as is

revealed by much lower TCS (16.333) and the absence of damage at class 3 and 4 levels. This

was the concentration that produced 50% inhibition of the axenically grown amastigotes of

Leishmania tropica as shown in chapter 3.0. Actinomycin D caused

Page 117: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

95

Figure 5.5. Genotoxicity of Actinomycin D (M1) at 100μM, 25μM and 2.135μM concentration

(which showed notable IC50 value for Leishmania tropica amastigotes in Chapter 3.0).

significant level of genotoxicity at 100 and 25μM concentrations on human lymphocytes as

compared to standard Miltefosine, 1% DMSO and negative control as well as Isosteviol

derivatives (P<0.05). At 2.135μM, however, the genotoxicity was non-significant in comparison

to the standard drug and 1% DMSO control as well as Isosteviol derivatives (P>0.05) but was

still significant as compared to negative control. Thus, concentration effective against the

parasite caused minimum and probably repairable genotoxic effects on the human PBLs.

5.3.6. Genotoxicity of Actinomycin Z3 (M2)

Actinomycin Z3 also showed a concentration-based genotoxicity in human peripheral

blood lymphocytes. It was highest at 100μM concentration (figure 5.6 A) followed by 25μM

(figure 5.6 B) and was minimum at 1.76μM (figure 5.6 C). The TCS of 302.33, 213, and 29.66

respectively were recorded which shows steep decrease with the reducing concentration of

compound used. The damage wrecked to class 1 and 2 only at the lowest concentration as is

revealed in figure 5.6-C. At the higher concentrations, class 1 and 2 damage was, however, less

as compared to the class 3 and 4 which occurred at greater rate (figure 5.5-A and B). In the

nutshell, the 50% inhibitory concentration of the compound was found with minimum genotoxic

Page 118: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

96

associations to the human peripheral blood lymphocytes. The compound was, however, found

causing significant genotoxicity as compared to Miltefosine, 1% DMSO, negative control and

Isosteviol derivative at all concentrations (P<0.05).

Figure 5.6. Genotoxicity of Actinomycin Z3 (M2) at 100μM, 25μM and 1.76μM concentration

(which showed notable IC50 value for Leishmania tropica amastigotes in Chapter 3.0).

5.3.7. Genotoxicity of Actinomycin Z5 (M3)

Actinomycin Z5 showed similar representation of genotoxicity on human blood lymphocytes

like the two other actinomycin family of compounds used. It was however, found more

genotoxic as compared to the Miltefosine. As is revealed in figure 5.7, genotoxicity experienced

a steep decline with the decrease in concentration. At 100μM highest value (337.66) of TCS was

noted and greater proportion of damage was represented at class 3 and 4 levels (figure 5.7-A). At

25μM, still enough genotoxic effects were caused on the lymphocytes with still a higher value of

TCS (214.33) and most of the damage represented at class 2, 3 and 4 levels (figure 5.7-B).

Genotoxic intensity, however, came to the minimum at 1.691μM with much lower TCS (23.666)

Page 119: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

97

and damage at the level of only class 1 and 2 levels being present. The compound showed

significant genotoxicity as compared to standard Miltefosine, 1% DMSO and negative control at

all the concentrations used in present investigation (P<0.05).

Figure 5.7. Genotoxicity of Actinomycin Z5 (M3) at 100μM, 25μM and 1.691μM concentration

(which showed notable IC50 value for Leishmania tropica amastigotes in Chapter 3.0).

5.4. Discussion

Genotoxic effects of an agent depend on the exposure time and concentration. They show a

direct proportionality to both the time and concentration. As assessed in vitro on human

leukocytes and in mice, exposure to 0.25, 0.5 and 1μg/mL of Miltefosine for a few hours caused

no significant damage but it became significant when exposure time increased to 48 hours

(Branco et al., 2016). In the present study, however, the drug was found less genotoxic to human

peripheral blood lymphocytes at the same exposure time at the lowest concentration. At higher

concentrations, however, genotoxic damage to the level of class 1 was gripped but not with

higher ratio.

Page 120: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

98

There has been evidence of associated genotoxicity regarding actinomycin which is a DNA

mediated RNA synthesis inhibitor (Daza et al., 2002). The same research group (Daza et al.,

2004), also reported actinomycin-D causing significant DNA damage at 10, 20 and 40μM

concentrations. A similar dose dependant genotoxicity was also recorded in gold fish (Carassius

auratus) skin primary cell culture exposed to actinomycin D (Ishikawa et al., 2005). Results of

our study are in consensus with the findings of these workers at the highest concentrations. At

the lowest concentration, however, different results were obtained as damage up to the level of

class 2 could be seen in this study. Actinomycin D, when used to check genotoxicity on

axenically grown corneal cells, showed dose dependant increase in the genotoxicity causing

highest damage at a concentration of 100μM (Sakaki et al., 2015). Actinomycin D has been

found to cause damage to DNA at 0.05μg/mL with comet tails measurement up to 30μm in

length (Kwaguchi et al., 2010). As revealed in the present study, actinomycin caused longer tails

in the lymphocytes showing consensus with the aforementioned study.

Isosteviol has been found to inhibit the activity of mammalian (including human), polymerase

and topoisomerase II, that play important role in DNA replication (Mizushina et al., 2005).

Stevioside and rebaudioside A, Isosteviol close kin metabolites, have been found to cause no

mutagenesis in certain studies. A few studies, however, like that of Nunes et al. (2007), have

found stevioside to cause genotoxicity at 4 mg/mL as evaluated by comet assay (Brusick, 2008;

Momtazi-Borojeni et al., 2016). At very higher dose i.e., 10 folds the recommended one, it has

been found to cause mutagenicity in bacteria (Suttajit et al., 1993). In other studies, stevioside

have been proved not to be mutagenic but steviol, a more related compound to Isosteviol, was

found to cause dose dependant mutagenic effects in bacteria and Chinese hamster lung fibroblast.

In the latter it also caused chromosomal aberrations (Matusi et al., 1996). A similar dose related

mutagenic response has also been confirmed by Pezzuto et al. (1985) in bacteria through forward

mutation assay. In the present study the Isosteviol derivatives were found to cause DNA

fragmentation in a dose dependant manner. At the highest concentration, all classes of damage

were seen while damage decreased to a minimum and class 01 at the lowest concentration. From

the present results, it can be concluded that chemical modification of Isosteviol and other

compounds from the diterpenoid groups, along with having antimicrobial effectiveness, may also

increase genotoxic and cytotoxic effects on pathogenic protozoa like Leishmania tropica.

Page 121: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

99

Chapter 6. General Discussion

There is an imperative need to develop novel treatment to combat leishmaniasis, as majority of

the existing medications are highly toxic, present resistance issues and are financially

unaffordable by poor victims. If we identify the intricate life-cycle phases of Leishmania and

mechanism of drug action, then our capability to control leishmaniasis will advance. In the

present investigation variances in pathogenicity, transmittable prospectives and their response to

antileishmanial compounds have been studied. In previous studies, temporal morphological

variability of Leishmania species during life cycle stages was measured with specific metabolic,

physiologic, structural, surface protein and genetic differences (Wheeler et al., 2011).

Predisposed by such differences is variability in pathogenicity, infective potential, vector

specificity and response to antileishmanial agents of these life cycle stages. There have been

several detailed studies to evaluate morphologically variable forms in sand fly vector (Killick-

Kendrick et al., 1996; Gossage et al., 2003; Kamhavi, 2006; Frietas et al., 2012). There are,

however, fewer studies to evaluate phenotypically variable forms in vitro. Such assessment

during axenic cultivation of Leishmania is indispensable prior to experimental infection in model

animals, isolated macrophages and drug sensitivity assays. Recently there have been

improvement of more sensitive tools to select infective metacyclic promastigotes, but they have

associated limitations including requirement of expensive apparatuses, lengthy protocols and

histochemical and molecular preparations (Calcagno et al., 2002). This renders morphological

association of different stages during in vitro cultivation an important task regarding Leishmania

laboratory studies. Sorting of temporally transitional promastigote stages in axenic growth

through microscopy has been performed throughout and is still exercised. Careful follow up

usually ends up with better, if not best, results regarding metacyclic selection for further use.

Light microscopic analysis taken jointly with scanning electron microscopy, can bring precise

correlation of different stages of promastigotes (Chang, 1979). A greater move is seen towards

morphological and morphometric approaches, equalling histochemical and molecular one, for

ready and easy morphotype assessment in axenic cultivation. Axenically grown amastigotes

provide a substitute for intramacrophage stage of the parasite. Different species of Leishmania

require different conditions to transform into amastigotes (Wheeler et al., 2011; Chanmol et al.,

2019). During the present study Leishmania tropica was successfully transformed to amastigotes

Page 122: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

100

in culture by just leaving promastigotes in culture without medium changes. It was found that,

while transforming, parasites are not getting completely round but many remain ovoid. Loss of

extracellular flagellum and reduction in cell size were shown to be the main criteria to determine

that transformation has occurred as determined previously by Ismaeel et al. (1998).

Cytocidal effects of an agent in candidature for therapeutics in an infection must be evaluated

before its use in vivo. In vitro studies on both the normal and malignant cells provide a good

model for such assessments and are often crucial in biomedical findings and designing drug

administration and dosage schedules (Valeriote et al., 1973; Akoi et al., 1998). Blood is the

immediate exposed body tissue to any material that enters the body. Blood cells, therefore, are

convenient entities for in vitro chemical and pharmaceutical testing (Andersson et al., 2003;

Faust et al., 2004; Vorobyeva et al., 2007). Findings of the present study may be helpful in future

in vitro as well as in vivo studies on Isosteviol derivatives and actinomycins regarding their use

as antileishmanial agents. Dosage schedules with sub lethal concentrations of these agents may

be designed based on the current finings. In correlation to the current cytotoxic values, future

animal trials may be planned regarding use of Isosteviols and actinomycins against cutaneous

and visceral forms of leishmaniasis.

The re-emergence of leishmaniasis has provoked concerns amongst the researchers to find new

and safer therapeutics for the infection (Mendes et al., 2016). Natural substances have been of

special concern in treating various ailments. Diterpenoids have been used in folkloric medicine

for several different conditions and are now gaining greater attention from researchers

(Nagarajan and Brindha, 2012; Strobykina et al., 2015). Actinomycins are known for their

antimicrobial and anti-neoplastic activities since 1950s. Actinomycin D is the most familiar of

the group (Alenazi et al., 2018). It has shown potential in vitro antiprotozoal activity against

Plasmodium falciparum strain 3D7 (Fink and Goldenberg, 1969). It also has been found to cause

the prevention of heat shock proteins in Leishmania major promastigotes (Lawrence and Robert-

Gero, 1985). However, there has been no comprehensive study of both the diterpenoids and

actinomycins against Leishmania tropica. The present study may therefore be amongst the

pioneering efforts to evaluate antileishmanial effects of diterenoid Isoteviol analogues and

actinomycins. This will provide baseline information regarding the antileishmanial potency of

these candidates. Based on the findings of the current attempt, actinomycins may be of prime

Page 123: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

101

significance against cutaneous infection caused by Leishmania tropica. On the other hand, this

investigation may offer a solid base for the strengthening of other antiprotozoal studies and in

principally how other protozoan parasites react to actinomycins and diterenoid Isoteviol

analogues in vitro.

Comet assay is one of the several techniques used to assess DNA damage and mutagenesis

which result in a variety of different conditions including carcinogenesis and teratogenesis (Tice

et al., 2000; Pereira et al., 2012). Comets are generally visualized by using fluorescent dye

technique that need fluorescent microscopy which is expensive device and not affordable in low

budget researches. Giemsa stain has been evaluated by just one earlier study (Osipov et al.,

2014) as a cheaper and effective alternative for comet visualization. The present study supports

the original study positively. We visualized all classes of comets using Giemsa stain. The results

were not less than where comets were visualized with fluorescent techniques. Use of Giemsa

staining technique for comet assay will probably be adopted as an inexpensive readily available

alternative. Careful and expert handling of the stain is expected to bring comparable, if not

superior, results to fluorescent approaches. Use of fresh and double filtered 10% working

Giemsa stain solution has sown greater clarity. Based on the present Comet assay results,

actinomycins, in higher doses, can be used as positive control for genotoxicity studies in future.

Page 124: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

102

Conclusion

Leishmania tropica (MHOM/PK/2010/KWH23) grows satisfactorily in RPMI-1640 medium

achieving a density of 1x107/mL in late log phase. Greater density of metacyclic forms is seen

towards the stationary phase which is achieved after 7 days of growth. Metacyclics are

characterized by their specific carrot tuber or cone shaped morphology with broader anterior and

much narrower posterior end. Loss of extracellular flagellum must be used as criterion for

identification of axenic amastigote. Orthophosphoric acid works satisfactorily for medium

acidication for axenic amastigotes as it is not volatilized at 37ᴼC to the free space in culture

flask. Isosteviol derivatives 16 (2,4-dinitrophenylhydrazine) Isosteviol, 17-hydroxy 16 (2,4-

dinitrophenylhydrazine) Isosteviol and Benzyl ester 16 (2,4-dinitrophenylhydrazine) Isosteviol

showed less cytotoxicity and genotoxicity than actinomycin D, Z3 and Z5 in vitro on human

peripheral blood lymphocytes. Genotoxic activity of both the classes of compounds is dependent

on concentration. Actinomycins, however, carry much higher genotoxic characteristics which

may be probable cause of their limited use as therapeutics. Both the classes of compounds, based

on the current in vitro findings, may be potential antileishmanial agents.

Page 125: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

103

Recommendations

Considering the present findings, following suggestions are put forward for researchers to

follow;

1. Extensive investigations of actinomycins against Leishmania are needed in vivo in animal

models with broader range of dose regimens and protocols using oral, intramuscular,

intravenous, intraleisional and topical drug delivery routes.

2. Evaluation of actinomycins through intraleisional and topical formulations shall be of

prime concern against cutaneous species of Leishmania. Use of drugs through topical

route may have less probable side effects due to less amount of drug used and by not

bringing drug in direct contact with more delicate internal organs and systems.

3. In vivo evaluation of cytotoxicity, genotoxicity and teratogenicity of actinomycins, again

with broader spectrum of treatment regimen, shall be conducted in association to

Leishmania infection.

4. Establishment of finer optical protocols for morphologic and morphometric analysis of

promastigote morphotypes would be highly appreciable as it provides ready to use

approach. Providing such data for greater number of species and strains is of immediate

need.

5. Derivatives of Isosteviol with a myriad of functional groups may provide antileishmanial

candidates in future studies.

6. In vitro transformation to amastigote is needed to be evaluated further for genotypic and

chromosomal variability and dividing ability.

Page 126: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

104

References

Abdeladhim, M., Kamhawi, S. and Valenzuela, J.G. (2014). What’s behind a sand fly bite? The

profound effect of sand fly saliva on host hemostasis, inflammation and immunity.

Infection, Genetics, and Evolution, 28: 691–703.

Abdossamadi, Z., Seyed, N. and Rafati, S. (2016). Mammalian host defense peptides and their

implication on combating Leishmania infection. Cellular Immunology, 309: 23-31.

Adl, S.M., Simpson, A.G., Lane, C.E, Lukeš, J., Bass, D., Bowser, S.S., et al. (2102). The

revised classification of eukaryotes. Journal of Eukaryote Microbiology, 59:429–93.

Ajdary, S., Jafari-Shakib, R., Riazi-Rad, F. and Khamesipour, A. (2009). Soluble CD26 and

CD30 levels in patients with anthroponotic cutaneous leishmaniasis. Journal of

Infection, 55: 75-78.

Ajdary, S., Riazi-Rad, F., Alimohammadian, M. and Pakza, S. (2009). Immune response to

Leishmania antigen in anthroponotic cutaneous leishmaniasis. Journal of Infection,

59:139-143.

Akhoundi, M., Khuls, K., Cannet, A., Votýpka, J., Marty, P., Delaunay, P. and Sereno, D.

(2016). A historical overview of the classification, evolution and dispersion of

Leishmania parasite and sand flies. Plos Neglected Tropical Diseases, DOI:

10.1371/journal.pntd.0004349.

Akhtar, M.S., Degaga1and, B. and Azam, T. (2014). Antimicrobial activity of essential oils

extracted from medicinal plants against the pathogenic microorganisms: A review.

Issues in Biological Sciences and Pharmaceutical Research, 2 (1): 1-7.

Akhtari, J., Faridnia, R., Kalani, H., Bastani, R., Fakhar, M., Rezvan, H.and Beydokhti. A.K.

(2019). Potent in vitro antileishmanial activity of a nanoformulation of cisplatin with

carbon nanotubes against Leishmania major. Journal of Global Antimicrobial

Resistance, 16: 11-16

Akoi, M., Kuwamura, M., Kotani, T., Katamoto, H., Kubo, K., Nomura, K., Sasaki, N. and

Ohashi, F. (1998). In vitro cytotoxicity of recombinant human TNF-α and actinomycin-

D on canine normal and tumour cells. Journal of Veterinary and Medical Sciences, 60

(10):1087-1091.

Alali, F.Q., Amrine, C.S.M., El-Elimat, T., Alkofahi, A., Tawaha, K., Gharaibah, M., Swanson,

S.M., Falkinham, J.O., Cabeza, M., Sánchez, A., Figueroa, M. and Oberlies, N.H.

Page 127: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

105

(2014). Bioactive withanolides from Withania obtusifolia. Phytochemistry Letters, 9:

96–101.

Al-Dhabi, N.A., Arasu, M.V. and Rejiniemon, T.S. (2015). In vitro antibacterial, antifungal,

antibiofilm, antioxidant, and anticancer properties of isosteviol isolated from

endangered medicinal plant Pittosporum tetraspermum. Evidence-Based

Complementary and Alternative Medicine Volume 2015, Article ID 164261, 11 pages

http://dx.doi.org/10.1155/2015/164261

Alenazi, F.S.H., Ahmed, M.Q., Fazaludeen, M.F., Shahid, S.M.A. and Kausar, M.A. (2018).

Actinomycin used as a potent inhibitor for malaria parasite Plasmodium Falciparum.

International Journal of Pharmaceutical and Phytopharmacological Research, 8 (2):

40-44.

Andersson, M., Agurell, E., Vaghef, H., Bolcsfoldi, G. and Hellman, B. (3003). Extended-term

cultures of human T-lymphocytes and the comet assay: a useful combination when

testing for genotoxicity in vitro? Mutation Research, 540 (2003) 43–55.

Andrade, B.B., De Oliveira, C.I., Brodskyn, C.I., Barral, A. and Barral-Neto, M. (2007). Role of

sand fly saliva in human and experimental leishmaniasis: current insights. Scandinavian

Journal of Immunology, 66 (2-3): 122-127.

Ang, M.L.T, Murima, P. and Pethe, K. (2015). Next-generation antimicrobial: from chemical

biology to first-in-class drugs. Archives of Pharmaceutical Research, 38: 1702-1717.

Anthony, B. M. & Helmut, L. (2011). The actinomycins in Anticancer agents from natural

products. Second Edition 363–382 (CRC Press, London).

Aoki M., Kuwamura, M., Kotyani, T., Kotamoto, H., Kobo, K., Nomura, K., Sasaki, N. and

Ohashi., F. (1998). In vitro cytotoxicity of recombinant human TNF-α and actinomycin

Don canine normal and tumor cells. Journal of Veterinary and Medical Sciences, 60

(10). 1087-1091.

Ashutosh, Sundar, S. and Goyal, N. (2007). Molecular mechanism of antimony resistance in

Leishmania. Journal of Medical Microbiology, 56: 143-153.

Asilian, A., Jalayer, T., Nilforooshzadeh, M., Ghassemi, R.L., Peto, R., Wayling, S., Olliaro, P.

and Modabber, F. (2003). Treatment of cutaneous leishmaniasis with aminosidine

(paromomycin) ointment: double-blind, randomized trial in the Islamic Republic of

Iran. Bulletin of the World Health Organization, 81 (5): 353-359.

Page 128: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

106

Atanasov, A.G., Waltenberger, B., Pferschy-Wenzig, E., Linder, T., Wawrosch, C., Uhrin, P.,

Temml, V., Wang, L., Schwaiger, S., Heiss, E.H., Rollinger, J.M., Schuster, D., Breuss,

J.M., Bochkov, V., Mihovilovic, M.D., Kopp, B., Bauer, R., Dirsch, V.M. and Stuppner,

H. (2015). Discovery and resupply of pharmacologically active plant-derived natural

products: A review. Biotechnology Advances, 33: 1582–1614.

Atia, A.M., Mumina, A., Tayler-Smith, K., Boulle, P., Alcoba, G., Elhag, M.S., Alnour, M.,

Shah, S., Chappuis, F., van Griensven, J., and Zachariah, R. (2015). Sodium

stibogluconate and paromomycin for treating visceral leishmaniasis under routine

conditions in eastern Sudan. Tropical Medicine and International Health, 20 (12):

1674-1684.

Auon, K. and Bouratbine, A. (2014). Cutaneous leishmaniasis in North Africa: a review.

Parasite, 21: 14.

Ayres, D. C., Pinto, L. A. and Giorgio, S. (2008). Efficacy of pentavalent antimony,

amphotericin B, and miltefosine in Leishmania amazonensis-infected macrophages

under normoxic and hypoxic conditions. Journal of Parasitology, 94 (06): 1415-1417.

Azanza, J.R., Sádaba, B. and Reis, J. (2015). Liposomal formulations of amphotericin B:

differences according to the scientific evidence. Revista Espanola de Quimioterapia, 28

(6): 275-281.

Bahrami, S., Hatam, G.R., Razavi, M. and Nazifi, S. (2011). In vitro cultivation of axenic

amastigotes and the comparison of antioxidant enzymes at different stages of

Leishmania tropica. Tropical Biomedicine, 28 (2): 411–417.

Balaña-Fouce, R., Reguera, R.M., Cubría, J. C. and Ordóñez, D. (1998). The pharmacology of

leishmaniasis. General Pharmacology, 30 (4): 435–443.

Balaña-Fouce. R.., Ordóñez, D. and Alunda, J. M. (1989). Putrescine transport system in

Leishmania infantum promastigotes. Molecular and Biochemical Parasitology, 35: 43–

50.

Balasegaram, M., Ritmeijer, K., Lima, M.A., Burza, S., Genovese, G.O., Milani, B., Gaspani, S.,

Potet, J. and Chappuis, F. (2012). Liposomal amphotericin B as a treatment for human

leishmaniasis. Expert Opinion. Emerging Drugs, 17 (4): 493-510.

Banerjee, A., De, M.and Ali, N. (2011). Combination therapy with paromomycin-associated

stearylamine-bearing liposomes cures experimental visceral leishmaniasis through th1-

Page 129: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

107

based immunomodulation. Antimicrobial Agents and Chemotherapy, 55 (4): 1661–

1670.

Bari, I. and Rahman, S. (2008). Cutaneous leishmaniasis: an overview of parasitology and host-

parasite-vector inter-relationship. Journal of Pakistan Association of Dermatologists,

18: 42-48.

Batista, R., García, P.A., Castro, M.A., del Corral, J.M.M, Speziali, N.L., Varotti, F.deP., de

Paula, R.C., García-Fernández, L.F., Francesch, A., Feliciano, A.S. and de Oliveira,

A.B. (2013). Synthesis, cytotoxicity and antiplasmodial activity of novel ent-kaurane

derivatives. European Journal of Medicinal Chemistry, 62: 168-176.

Batista, R., Humberto, J.L., Chiari, E. and de Oliveira, A.B. (2007) Synthesis and trypanocidal

activity of ent-kaurane glycosides. Bioorganic and Medicinal Chemistry, 15: 381-391.

Bavarsad, N., Bazzaz, B.S.F., Khamesipour, A. and Jaafari, M.R. (2012). Colloidal, in vitro and

in vivo anti-leishmanial properties of transfersomes containing paromomycin sulfate in

susceptible BALB/c mice. Acta tropica, 124: 33-41.

Beattie, L. and Kaye, P.M. (2011). Leishmania-host interactions: what has imaging taught us?

Cellular Microbiology, 13 (11). 1959-1967.

Bee, A., Culley F.J., Alkhalife, I.S., Bodman-Smith, K.B., Raynes, J.G. and Bates, P.A. (2001).

Transformation of Leishmania mexicana metacyclic promastigotes to amastigote-like

forms mediated by binding of human C-reactive protein. Parasitology, 122: 521–529.

Beer, M.F., Frank, F.M., Elso, O.G., Bivona, A.E., Cerny, N., Giberti, G., Malchiodi, E.L.,

Martino, V.S., Alonso, M.R., Sülsen, V.P. and Cazorla, S.I. (2016). Trypanocidal and

leishmanicidal activities of flavonoids isolated from Stevia satureiifolia var.

satureiifolia. Pharmaceutical Biology, 54 (10): 2188-2195. DOI:

10.3109/13880209.2016.1150304

Bell, C.A., Hall, J.E., Kyle, D.E., Grogl, M., Ohemeng, K.A., Allen, M.A. and Tidwell, R.R.

(1990). Structure-activity relationships of analogs of pentamidine against Plasmodium

falciparum and Leishmania mexicana amazonensis. Antimicrobial Agents

and Chemotherapy, 34:1381-1386.

Bérdy, J. (2005). Bioactive microbial metabolites. Journal of Antibiotics, 58 (1): 1-26.

Bezerril, A.B. and Teixeira, C.R. (2012). Biomarkers for exposure to sand flies bites as tools to

aid control of leishmaniasis. Frontiers in Immunology, 3:121-7

Page 130: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

108

Birkholtz, L., Williams, M., Niemand, J., Louw, A.I., Persson, L. and Heby, O. (2011).

Polyamine homoeostasis as a drug target in pathogenic protozoa: peculiarities and

possibilities. Biochemical Journal, 438: 229-244.

Bogdan, C. (2012). Natural killer cells in the experimental human leishmaniasis. Frontiers in

Cellular and Infection Microbiology, 2: doi: 10.3389/fcimb.2012.00069.

Bogdan, C. and Röllinghoff, M. (1998). The immune response to Leishmania: mechanism of

parasite control and evasion. International Journal for Parasitology, 28: 121-134.

Brahim, M.A.S., Fadli, M., Hassani, L., Boulay, B., Markouk M., Bekkouche, K., Abbad, A.,

Ali, M.A. and Larhsini, M. (2015). Chenopodium ambrosioides var. ambrosioides used

in Moroccan traditional medicine can enhance the antimicrobial activity of conventional

antibiotics. Industrial Crops and Products, 71: 37–43.

Branco, P.V.C.; Soares, R.P.; de Jesus, L.C.L.; Moreira, V.R.; Alves, H.J.; Belfort, M.R.deC.;

Silva, V.L.M. and Pereira, SRF. (2016). The antileishmanial drug miltefosine

(Impavido®) causes oxidation of DNA bases, apoptosis, and necrosis in mammalian

cells. Mutation Research, 806: 34–39.

Brochu, C., Wang, J., Roy, G., Messier, N., Wang, X., Saravia, B.G., and Ouellette, M. (2003).

Antimony uptake systems in the protozoan parasite Leishmania and accumulation

differences in antimony-resistant parasites. Antimicrobial Agents and Chemotherapy, 47

(10): 3073-3079.

Brusick, D.J. (2008). A critical review of the genetic toxicity of steviol and steviol glycosides.

Food and Chemical Toxicology, 46: 83–91.

Burza, S., Croft, S. L and Boelaert, M. (2018). Leishmaniasis. Lancet, 392 (10151): 951-970.

doi: 10.1016/S0140-6736(18)31204-2

Caballero, A.B., Salas, J.M. and Sánchez-Moreno, M. (2015). Metal-based therapeutics for

leishmaniasis. http://dx.doi.org/10.5772/57376.

Calcagno, M., Avilan, L., Colasante, C., Berrueta, L. and Salmen, S. (2002). Interaction of

different Leishmania mexicana morphotypes with plasminogen. Parasitology Research,

88: 972–978. DOI 10.1007/s00436-002-0688-2

Cantacessi, C., Dentas-Torres, F., Nolan, M.J. and Otranto, D. (2015). The past, present and

future of Leishmania genomics and transcriptomics. Trends in Parasitology, 31 (03):

100-108.

Page 131: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

109

Carneiro, P.P., Conceição, J., Macedo, M., Magalhães, V., Carvalho, E.M. and Bacellar, O.

(2016). The role of nitric oxide and reactive oxygen species in the killing of Leishmania

braziliensis by monocytes from patients with cutaneous leishmaniasis. PLoS ONE, 11

(2): e0148084. doi:10.1371/journal.

Carriόn, J., Folguerira, C., Soto, M., Frenso, M., Requena, J.M. (2011). Leishmania infantum

HSP70-II null mutant as candidate vaccine against leishmaniasis: a preliminary

evaluation. Parasites and vectors, 4: 150.

Cherney, E.C., Green, J.C. and Baran, P.S. (2013). Synthesis of ent-kaurane and beyerane

diterpenoids via controlled fragmentations of overbred intermediates. Angewandte

Chemie International Edition in English, 52 (34): doi:10.1002/anie.201304609.

Cooper, H.L.; Braverman, R. (1977). The mechanism by which actinomycin D inhibits protein

synthesis in animal cells. Nature, 269: 527-529.

Corrêa, J.R., Brazil, R.P. and Soares, M.J. (2005). Leishmania (Viannia) lainsoni

(Kinetoplastida: Trypanosomatidae), a divergent Leishmania of the Viannia subgenus –

A mini review. Memõrias do Instituto Oswaldo Cruz, 100 (6): 587-592.

Costa, M.S., Gonçalves, Y.G., Nunes, D.C.O., Napolitano, D.R., Maia, P.I.S., Rodrigues, R.S.,

Rodrigues, V.M., Poelhsitz, G.V. and Yoneyama, K.A.G. (2017). Anti-Leishmania

activity of new ruthenium (II) complexes: Effect on parasite-host interaction. Journal of

Inorganic Biochemistry, doi: 10.1016/j.jinorgbio.2017.07.023

Cox, F.E.G. (2002). History of human parasitology. Clinical Parasitology Reviews, 15 (04): 595-

612.

Croft, S.L. and Coombs, G.H. (2003). Leishmaniasis– current chemotherapy

and recent advances in the search for novel drugs. TRENDS in Parasitology, 19 (11):

doi:10.1016/j.pt.2003.09.008.

Croft, S.L. and Olliaro, P. (2011). Leishmaniasis chemotherapy—challenges and opportunities.

Clinical Microbiology and Infection. 10.1111/j.1469-0691.2011.03630. x.

Croft, S.L., Seifert, K. and Yardley, V., 2006. Current scenario of drug development for

leishmaniasis. Indian Journal of Medical Research, 123, 399–410.

Cupolillo, E., Medina-Acosta, E., Noyes, H., Momen, H. and Grimaldi Jr, G. (2000). A revised

classification for Leishmania and Endotrypanum. Parasitology Today, 16 (04): 142-144.

Page 132: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

110

Cysne-Finkelstein, L., Temporal, R.M., Alves, F.A. and Leon, L.L. (1998). Leishmania

amazonensis: long-term cultivation of axenic amastigotes is associated to

metacyclogenesis of promastigotes. Experimental Parasitology, 89: 58–62.

Da Costa FB, Vichnewiski W, Herz W. 1996. Diterpenes and synthetic derivatives from

Viguiera aspillioides with trypanomicidal activity. Planta Medica, 62: 557–559.

da Cruz, M.Q., Bräscher, H.M., Vargens, J.R., Maia, L.C. and Oliveira-Lima, A. (1989).

Absence of immunosuppression in DBA/2 mice vaccinated with Trypanosoma cruzi

treated with actinomycin-D. Experientia, 45 (11-12):1131-33.

da Rosa, R., de Moraes, M.H., Zimmermann, L.A., Schenkel, E.P., Steindel, M., Sibelle, L. and

Bernardes, C. (2017). Design and synthesis of a new series of 3,5-disubstituted

isoxazoles active against Trypanosoma cruzi and Leishmania amazonensis. European

Journal of Medicinal Chemistry, 128: 25-35.

Da Silva, R. P., and Sacks, D. L. 1987. Metacyclogenesis is a major determinant of Leishmania

promastigote virulence and attenuation. Infection and Immunity, 55: 2802–2807.

Daza, P.; Torreblanca, J. and Moreno, F.J. (2004). The comet assay differentiates efficiently and

rapidly between genotoxins and cytotoxins in quiescent cells. Cell Biology

International, 28: 497-502.

Daza, P.; Torreblanca, J.; Garcı´a-Herdugo, G. and Moreno, F.J. (2002) DNA strand breaks

induced by the topoisomerase I inhibitor camptothecin in unstimulated human white

blood cells. Cell Biology International, 6 (8): 707-13.

de Alencar, M.V.O.B., de Sousa, J.M.deC., Rolim, H.M.L., de Medeiros, M.dasG.F., Cerqueira,

G.S., Almeida, F.R.deC., Citó, A.M.dasG.L., Ferreira, P.M.P., Lopes, J.A.D., Melo-

Cavalcante, A.A.deC. and Islam, M.T. (2017). Diterpenes as lead molecules against

neglected tropical diseases. Phytotherapy Research, 31: 175-201.

de Figueiredo, A.B., Souza-Testasicca, M.C. and Afonso, L.C.C. (2016). Purinergic signaling

and infection by Leishmania: A new approach to evasion of the immune response.

Biomedical Journal, 39: 244-250.

de Morais-teixeira, E., Damasceno, Q.S., Galuppo, M.K., Romanha, A.J. and Rabello, A. (2011).

the in vitro leishmanicidal activity of hexadecylphosphocholine (miltefosine) against

four medically relevant Leishmania species of Brazil. Memõrias Instituto Oswaldo

Cruz, 106 (4): 475-478.

Page 133: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

111

de Santana, F.R., Dalboni, L.C., Nascimento, K.F., Konno, F.T., Alvares-Saraiva, A.M., Correia,

M.S.F., Bomfim, M.D.C., Casarin, R.C.V., Perez, E.C., Lallo, M.A., Peres, G.B.,

Laurenti, M.D., Benites, N.R., Buchi, D.F. and Bonamin, L.V. (2017). High dilutions of

antimony modulate cytokines production and macrophage – Leishmania (L.)

amazonensis interaction in vitro. Cytokine, 92: 33-47.

Debrabant, A., Joshi, M.B., Pimenta, P.F.P. and Dwyer, D.M. (2004). Generation of Leishmania

donovani axenic amastigotes: their growth and biological characteristics. International

Journal of Parasitology, 34: 205-217.

Demarchi, I.G., Thomazella, M.V., Terron, M.S., Lopes, L., Gazim, Z.C., Cortez, D.A.G.,

Donatti, L., Aristides, S.M.A., Silveira, T.G.V. and Lonardoni, M.V.C. (2015).

Antileishmanial activity of essential oil and 6,7-dehydroroyleanone isolated from

Tetradenia riparia. Experimental Parasitology, 157:128-137.

Der Auwera, G.V., Fraga, J., Montalovo, A.M. and Dujardin, J. (2011). Leishmania taxonomy up

for promotion? Trends in Parasitology, 27 (2): 49-50.

Dias, C.N., Rodrigues, K.A.F., Carvalho, F.A.A., Carneiro, S.M.P., Maia, J.G.S., Andrade,

E.H.A. and Moraes, D.F.C. (2013). Molluscicidal and leishmanicidal activity of the leaf

essential oil of Syzygium cumini (L.) Skeels from Brazil. Chemistry and Biodiversity,

10: 1133-1141.

Díaz, M.V., Miranda, M.R., Campos-Estrada, C., Reigada, C., Maya, J.D., Pereira, C.A. and

López-Muñoz, R. (2014). Pentamidine exerts in vitro and in vivo anti Trypanosoma

cruzi activity and inhibits the polyamine transport in Trypanosoma cruzi. Acta Tropica,

134: 1-9.

Diekmann-Schupert, A. and Franklin, R.M. (1990). Mode of action of tubolozole against

Plasmodium falciparum in vitro. Antimicrobial Agents and Chemotherapy, 34 (08):

1529-1534.

Diro, E., Ritmeijer, K., Boelaert, M., Alves, F., Mohammed, R., Abongomera, C., Raffaella

Ravinetto, R.., Crop, M.D., Fikre, H., Adera, C., Colebunders, R., Loen, H.V., Menten,

J., Lynen, L., Hailu, A. and Griensven, J.V. (2015). Use of pentamidine as secondary

prophylaxis to prevent visceral leishmaniasis Relapse in HIV infected patients, the first

twelve months of a prospective cohort study. PLoS Neglected Tropical Diseases, 9 (10):

e0004087. doi:10.1371/journal.pntd.0004087.

Page 134: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

112

Dong, M., Cao, P., Ma, Y., Luo, J., Yan, Y., Li, R. and Huang, S. (2018). A new actinomycin Z

analogue with an additional oxygen bridge between chromophore and β-

depsipentapeptide from Streptomyces sp. KIB-H714. Natural Product Research, DOI:

10.1080/14786419.2018.1443097

Dorlo, T.P.C., Balasegaram, M, Beijnen, J.H. and de Vries, P.J. (2012). Miltefosine: a review of

its pharmacology and therapeutic efficacy in the treatment of leishmaniasis. Journal of

Antimicrobial Chemotherapy, 67: 2576–2597.

Dostálová, A. and Volf, P. (2012). Leishmania development in sand flies: parasite-vector

interactions overview. Parasites & Vectors, 5:276.

Dougall, A.M., Alexander, B., Holt, D.C., Harris, T., Sultan, A.H., Bates, P.A., Rose, K. and

Walton, S.F. (2011). Evidence incriminating midges (Diptera: Ceratopogonidae) as

potential vectors of Leishmania in Australia. International Journal for Parasitology, 41:

571-579.

Duke, S.O., Dayan, F.E., Romagni, J.G. and Rimando, A.M. (2000). Natural products as sources

of herbicides: Current status and future trends. Weed Research, 40: 99-111.

Dykstra, C.C. and Tidwell, R. R. (1991). Inhibition of topoisomerases from Pneumocystis carinii

by aromatic dicationic molecules. Journal for Protozoology, 38: 78S-81S.

Eissa, M.M., Amer, E.I., Mossallam, S.F., Gomaa, M.M. and Baddour, N.M. (2012). Miltefosine

for Old World cutaneous leishmaniasis: An experimental study on Leishmania major

infected mice. Alexandria Journal of Medicine, 48, 261–271.

Evans, K.J. and Kedziersky, L. (2012). Development of vaccines against visceral leishmaniasis.

Journal of Tropical Medicine, Article ID 892817, 14 pages, doi:10.1155/2012/892817.

Fairlamb, A. H. and Cerami, A. (1992) Metabolism and function of trypanothione in the

kinetoplastida. Annual Review of Microbiology, 46: 695–729.

Faust, F., Kassie, F., Knasmüller, S., Boedecker, R.H., Mann, M. and Mersch-Sundermann, V.

(2004). The use of the alkaline comet assay with lymphocytes in human biomonitoring

studies. Mutation Research, 566: 209–229.

Fernandes, K.S., de Souza, P.E.N., Dorta, M.L. and Alonso, A. (2017). The cytotoxic activity of

miltefosine against Leishmania and macrophages is associated with dynamic changes in

plasma membrane proteins. Biochimica Biophysica Acta, 1859: 1–9.

doi: 10.1016/j.bbamem.2016.10.008

Page 135: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

113

Ferro, M and Doyle, A. (2001). Standardization for in vitro toxicity tests. Cell Biology and

Toxicology, 17 (4-5): 205-12.

Fiebig, M., Kelly, S. and Gluenz, E. (2015). Comparative life cycle transcriptomics

revises Leishmania mexicana genome annotation and links a chromosome duplication

with parasitism of vertebrates. PLOS Pathogens, DOI:10.1371/journal.ppat.1005186.

Fink, E. and Goldenberg, D.M. (1969). Antimalarial activity of actinomycin D and

cyclophosphamide. Proceedings of the Society for Experimental Biology and Medicine,

132 (1): 165-170.

Fraga, J., Montalvo, A.M., de Doncker, S., Dujardin, J. and der Auwera, G.V. (2010). Phylogeny

of Leishmania species based on heat-shock protein 70 gene. Infection, Genetics and

Evolution, 10: 238-239.

Fraga, J., Montalvo, A.M., der Auwera, G.V., Maes, I., Dujardin, J. and Requena, J.M. (2013).

Evolution and species discrimination according to the Leishmania heat-shock protein 20

gene. Infection, Genetics and Evolution, 18: 229–237.

Frézard, F., Demicheli, C. and Ribeiro, R.R. (2009). Pentavalent antimonials: new

perspectives for old drugs. Molecules, 14: 2317-2336.

Freitas, V.C., Parreiras, K.P., Duarte, A.P.M., Secundino, N.F.C. and Pimenta, P.F.P. (2012).

Development of Leishmania (Leishmania) infantum chagasi in its natural sand fly

vector Lutzomyia longipalpis. American Journal of Tropical Medicine and Hygiene,

86(4): 606-612.

Frézard, F., Demicheli, C. and Ribeiro, R.R. (2009). Pentavalent antimonials: new perspectives

for old drugs. Molecules, 14: 2317-2336.

Fuchs, Y and Steller, H. (2015). Live to die another way: modes of programmed cell death and

the signals emanating from dying cells. Nature Reviews Molecular Cell Biology,16:

329–344.

Gaeumann, E., Perlog, V. and Vischer, E. (1966). Actinomycin Z and method of producing the

same. United States Patent Office, 3,282.787.

Galluzzi, L., Vitale, I., Aronson, S.A., Abrams, J.M., Adam, D. and Agostinis, P. et al. (2018).

Molecular mechanisms of cell death: recommendations of the nomenclature committee

on cell death 2018. Cell Death and Differentiation, 25 (3): 486–541

Page 136: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

114

Gambino, D. and Otero, L. (2017). Design of prospective antiparasitic metal-based compounds

including selected organometallic cores. Inorganica Chimica Acta, doi:

http://dx.doi.org/10.1016/j.ica. 2017.07.068

Gannavaram S, Bhattacharya P, Ismail N, Kaul A, Singh R and Nakhasi HL (2016). Modulation

of innate immune mechanisms to enhance Leishmania vaccine induced immunity: role

of coinhibitory molecules. Frontiers Immunology, 7:187.

Garifullin, B.F., Strobykina, I.Y., Sharipova, R.R., Kravchenko, M.A., Andreeva, O.V.,

Bazanova, O.B. and Kataev, V.E. (2016). Synthesis and antituberculosis activity of the

first macrocyclic glycoterpenoids comprising glucosamine and diterpenoid isosteviol.

Carbohydrate Research, 431: 15-24.

Gaspar, M.M., Calado, S., Pereira, J., Ferronha, H., Correia, I., Castro, H., Tomás, A.M. and

Cruz, M.E.M. (2015). Targeted delivery of paromomycin in murine infectious diseases

through association to nano lipid systems. Nanomedicine: Nanotechnology, Biology,

and Medicine, 11: 1851–1860.

Georgiadou, S.P., Makaritsis, K.P. and Dalekos, G.N. (2015) Leishmaniasis revisited: current

aspects on epidemiology, diagnosis and treatment. Journal of Translational Internal

Medicine, 3 (2). DOI: 10.1515/jtim-2015-0002.

Geroldinger, G., Tonner, M., Hettegger, H., Bacher, M., Monzote, L., Walter, M., Staniek, K.,

Rosenau, T. and Gille, L. (2017). Mechanism of ascaridole activation in Leishmania.

Biochemical Pharmacology, 132: 48–62.

Glennie, N.D. and Scott, P. (2016). Memory T cells in cutaneous leishmaniasis. Cellular

Immunology, 309: 50–54.

Gomes, R., Cavalcanti, K., Teixeira, C., Carvalho, A.M., Mattos, P.S., Cristal, J.R., Muniz, A.C.,

Miranda, J.C., de Oliveira, C.I. and Barral, A. (2016). Immunity to Lutzomyia whitmani

saliva protects against experimental Leishmania braziliensis infection. PLoS Neglected

Tropical Diseases, 10 (11): e0005078. doi: 10.1371/journal.pntd.0005078.

Gossage, S.M., Rogers, M.E. and Bates, P.A. (2003). Two separate growth phases during the

development of Leishmania in sand flies: implications for understanding the life cycle.

International Journal for Parasitology, 33: 1027–1034.

Grand, R.J.; Chong, D.A.; Isselbacher, K.J. (1972). Intracellular processing of disaccharidases:

the effect of actinomycin D. Biochimica Biophysica Acta, 261: 341-352.

Page 137: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

115

Griensven, J.V., Gadisa, E., Aseffa, A., Hailu, A., Beshah, A.M. and Diro, E. (2016). Treatment

of cutaneous leishmaniasis Caused by Leishmania aethiopica: A Systematic Review.

PLoS Neglected Tropical Diseases, 10(3): e0004495. doi:10.1371/journal.

pntd.0004495.

Grimm, F., Brun, R., and Jenni, L. 1991. Promastigote infectivity in Leishmania infantum.

Parasitology Research, 77: 185–191.

Gupta, G., Oghumo, S. and Satoskar, A.R. (2013). Mechanism of Immune evasion in

leishmaniasis. Advances in Applied Microbiology, 82: 155-184.

Gupta, S. and Nishi. (2010). Visceral leishmaniasis: Experimental models for drug discovery.

Indian Journal of Medical Research, 133: 27-39

Hall, D.H., Gu, G., Garcia-Anoveros, J., Gong, L., Chalfie, M and Driscoll, M. (1997).

Neuropathology of degenerative cell death in Caenorhabditis elegans. Journal of

Neuroscience, 17, 1033–1045.

Hamil, R.J. (2013). Amphotericin B formulations: comparative review of efficacy and toxicity.

Drugs, 73 (9): 919-934.

Hamilton-Miller, J.M.T. (1973). Chemistry and biology of polyene macrolide antibiotics.

Bacteriological Reviews, 37 (20): 166-196.

Haraguchi, S.K., Silva, A.A., Vidotti, G.J., dos Santos, P.V., Garcia, F.P., Pedroso, R.B.,

Nakamura, C.V., de Oliveira, C.M.A. and da Silva, C.C. (2011). Antitrypanosomal

activity of novel benzaldehyde thiosemicarbazone derivatives from kaurenoic acid.

Molecules, (16): 1166-1180. `doi:10.3390/molecules16021166

Harkins, K.M., Schwartz, R.S., Cartwright, R.A. and Stone, A.C. (2016). Phylogenetic

reconstruction supports supercontinent origin for Leishmania. Infection, genetics and

Evolution, 38: 101-109.

Hatam, G.R., Bahrami, S., Razavi, S.M. and Oryan, A. (2013). Isoenzyme and ultrastructural

characterization of Leishmania tropica axenic amastigotes and promastigotes.

Parasitology Research, 112: 643–648.

Hollstein, U. (1974). Actinomycin: Chemistry and Mechanism of Action. Chemical Reviews, 74

(6): 625-652.

Page 138: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

116

Horta, M.F., Mendes, B.P., Roma, E.H., Noronha, F.S.M., Macêdo, J.P., Oliveira, L.S., Duarte,

M.M. and Vieira, L.Q. (2012). Reactive oxygen species and nitric oxide in cutaneous

leishmaniasis. Journal of Parasitology Research, doi:10.1155/2012/203818.

Huang, T., Chou, B., Lin, C., Weng, J., Chou, C., Yang, L. and Lin, S. (2014). Synthesis and

antiviral effects of isosteviol-derived analogues against the hepatitis B virus.

Phytochemistry, 99: 107–114.

Huang, T., Yang, C., Kuo, Y., Chang, Y., Yang, L., Chou, B., Lin, S. (2015). Synthesis and anti-

hepatitis B virus activity of C4 amide-substituted isosteviol derivatives. Bioorganic &

Medicinal Chemistry, 23: 720–728.

Iovannisci, D.M., Plested, C.P. and Moe, G.R. (2010). Evidence for rosettes as unrecognized

stage in the life cycle of Leishmania parasites. The Journal of Eukaryotic Microbiology,

57 (05): 405-414.

Ishikawa, T.; Ogata, S.; Okumura, K. and Taguchi, H. (2005) Detection of DNA damages

induced by five model chemicals in goldfish Carassius auratus cells using comet assay.

Cytologia, 70 (1): 59–64.

Ismaeel, A. Y., Garmson, J.G., Molyneux, D.H. and Bates, P.A. (1998) Transformation,

development and transmission of axenically cultured amastigotes of Leishmania

mexicana in vitro and in Lutzomyia longipalpis. American Journal of Tropical

Medicine and Hygiene, 59 (3): 421–425.

Iwu, M.M., Jackson, J.E. and Schuster, B.G. (1994). Medicinal plants in the fight against

leishmaniasis. Parasitology Today, 10 (2):65-68.

Kamhawi, S. (2006). Phlebotomine sand flies and Leishmania parasites: friends or foes?

TRENDS in Parasitology, 22 (9). 439-445.

Kamitori, S. and Takusagawa. F. (1994). Multiple Binding Modes of Anticancer Drug

Actinomycin D: X-ray, Molecular Modeling, and Spectroscopic Studies of d

(GAAGCTTC)2-Actinomycin D Complexes and Its Host DNA. Journal of American

Chemical Society, 116, 4154-4165.

Kataev, V.E., Strobykina, I.Y., Andreeva, O.V., Garifullin, B.F., Sharipova, R.R., Katz, E. and

Pugh, L.H. (1961). Variations on a theme: The actinomycins. Applied Microbiology, 9

(3): 263-266.

Page 139: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

117

Kaur, S., Chauhan, K. and Sachdeva, H. (2014). Protection against experimental visceral

leishmaniasis by immunostimulation with herbal drugs derived from Withania

somnifera and Asparagus racemosus. Journal of Medical Microbiology, 63: 1328–1338.

Kawaguchi, S.; Nakamura, T.; Yamamoto, A.; Honda, G. and Sasaki, Y.F. (2010). Is the comet

assay a sensitive procedure for detecting genotoxicity? Journal of Nucleic Acids, Article

ID 541050, 8 pages doi:10.4061/2010/541050.

Khademvatan, S., Gharavi, M.J., Rahim, F. and Saki, J. (2011). Miltefosine-induced apoptotic

cell death on Leishmania major and L. tropica Strains. Korean J Parasitology, 49

(1):17; DOI: 10.3347/kjp.2011.49.1.17.

Khan, M.N. Messenger, L.A., Wahid, S. and Sutherland, C. (2016). Phylogenetic position of

Leishmania isolates from Khyber Pakhtunkhwa province Pakistan. Experimental

Parasitology, 167: 61-66.

Khaybullin, R.N., Strobykina, I.Y., Dobrynin, A.B., Gubaydullin, A.T., Chestnova, R.V.,

Babaev, V.M. and Kataev, V.E. (2012). Synthesis and antituberculosis activity of novel

unfolded and macrocyclic derivatives of ent-kaurane steviol. Bioorganic & Medicinal

Chemistry Letters, 22: 6909–6913.

Khisroon, M., Gul, A.; Khan, A., Ali, N., Zaidi, F., Rasheed, S.B., Ahmadullah.

and Akbar, H. (2015). Comet assay-based DNA evaluation of fuel filling stations and

automobile workshops workers from Khyber Pakhtunkhwa province, Pakistan. Journal

of Occupational Medicine and Toxicology, 10:27. DOI 10.1186/s12995-015-0069-2

Killick-Kendrick, R., Killick-Kendrick, M., Tang, Y. and Bastien, P. (1996). Metacyclic

promastigotes of Leishmania in the salivary glands of experimentally infected

phlebotomine sandflies Parasite, 3, 55-60.

Kip, A.E., Schellens, J.H.M., Beijnen, J.H. and Dorlo, T.P.C. (2017). Clinical pharmacokinetics

of systemically administered antileishmanial drugs. Clinical Pharmacokinetics, DOI:

10.1007/s40262-017-0570-0.

Koba, M. and Konopa, J. (2005). Actinomycin D and its mechanism of action. Postepy Higieny i

Medycyny Doswwiadczalnej, 59: 290-298.

Kubo, I., Xu, Y. and Shimizu, K. (2004). Antibacterial activity of ent-kaurene diterpenoids from

Rabdosia rosthornii. Phytotherapy Researh, 18: 180–183.

Page 140: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

118

Kwakye-Nuako, G., Mosore, M., Duplessis, C., Bates, M.D., Puplampu, N., Mensah-Attipoe, I.,

Desewu, K., Afegbe, G., Asmah, R.H., Jamjoom, M.B., Ayeh-Kumi, P.F., Boakye, D.A.

and Bates, P.A. (2015). First isolation of a new species of Leishmania responsible for

human cutaneous leishmaniasis in Ghana and classification in the: Leishmania enriettii

complex. International Journal for Parasitology, 45: 679–684.

Lainson, R. and Shaw, J.J. (1987) Evolution, classification and geographical distribution, in The

Leishmaniases in Biology and Epidemiology Vol. 1 (Peters, W. and Killick-Kendrick,

R., eds), pp 1–120, Academic Press, London.

Lawrence, F. and Robert-Gero, M. (1985). Induction of heat shock and stress proteins in

promastigotes of Leishmania species. Proceeding of National Academy of Science, 82:

4414-4417.

Le, T.B., Beaufay, C., Nghiem, D.T., Mingeot-Leclercq, M. and Quetin-Leclercq, J. (2017). In

Vitro anti-leishmanial activity of essential oils extracted from Vietnamese plants.

Molecules, 22: 1071: doi:10.3390/molecules22071071.

Lei, S.M., Ramer-Tait, A.E., Dahlin-Laborde, R.R., Mullin, K. and Beetham, J.K. (2010).

Reduced hamster usage and stress in propagating Leishmania chagasi promastigotes

using cryopreservation and saphenous vein inoculation. Journal of Parasitology,

96:103– 108.

Lieke, T., Nylén, S., Eidsmo, L., Schmetz, C., Berg, L. and Akuffo, H. (2011). The interplay

between Leishmania promastigotes and human Natural Killer cells in vitro leads to

direct lysis of Leishmania by NK cells and modulation of NK cell activity by

Leishmania promastigotes. Parasitology, 138, 1898–1909.

Lin, L., Lee, L., Sheu, S. And Lin, P. (2004). Study on the stevioside analogues of

steviolbioside, steviol, and isosteviol 19-alkyl dmide dimers: synthesis and cytotoxic

and antibacterial activity. Chemical and Pharmaceutical Bulletin, 52 (9) 1117—1122.

Llanes, A., Restrepo, C.M., Vecchio, G.D. and Anguizola, F.J. (2015). The genome of

Leishmania panamensis: insights into genomics of the L. (Viannia) subgenus. Scientific

Reports, 5: 8550 DOI: 10.1038/srep08550.

Lohoff, M., Gessner, A., Bogdan, C. and Röllinghoff, M. (1998). Experimental murine

leishmaniasis and the Th1/Th2 cell concept. The Tokai Journal of Experimental

and Clinical Medicine, 23 (6), 347–50.

Page 141: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

119

Loiseau, P.M., Mbongo, N., Bories, C., Boulard, Y. and Craciunescu, D.G. (2000). In vivo

antileishmanial action of Ir-(Cod)-pentamidine tetraphenylborate on Leishmania

donovani and Leishmania Major mouse models. Parasite, 7: 103-108.

Loría-Cervera, E.N. and Andrade-Narváeza, F.J. (2014). Animal models for the study of

leishmaniasis immunology. Revista do Instituto de Medicina Tropical Săo Paulo, 56

(1):1-11.

Lourie, E.M. and Yorke, W., 1939. Studies in chemotherapy XXI. The trypanocidal

action of certain aromatic diamidines. Annals of Tropical Medicine and Parasitology,

33, 289–304.

Lowe, G., Droz, A.S., Vilaivan, T., Weaver, G.W., Tweedale, L., Pratt, J.M., Rock, P.,

Yardley, V. and Croft, S.L. (1999). Cytotoxicity of (2,2′:6′,2′′-Terpyridine)platinum(II)

Complexes to Leishmania donovani, Trypanosoma cruzi, and Trypanosoma brucei.

Journal of Medicinal Chemistry, 42 (6): 999-1006.

Lukeš, J., Mauricio, I.L., Gabriele Schönian, G., Dujardin, J., Soteriadou, K., Dedet, J., Kuhls.

K., Tintaya, K.W.Q., Jirku, M., Chocholová, E., Haralambous, C., Pratlong, F.,

Oborník, M., Horák, A., Ayala, F.J. and Miles, M.A. (2007). Evolutionary and

geographical history of the Leishmania donovani complex with a revision of current

taxonomy. PNAS, 104 (22): 9375-9380.

Lukeš, J., Skalický, T., Týč, J., Votýpka, J. and Yurchenko, V. (2014). Evolution of parasitism in

kinetoplastid flagellates. Molecular and Biochemical Parasitology, 195: 115–122.

Luketa, S. (2012). New views on the mega classification of life. Parasitology, 07 (04): 218-237.

Lundgren, G. and Moller, G. (1969. Non-specific induction of cytotoxicity in normal human

lymphocytes in vitro: studies of mechanism and specificity of the reaction. Clinical and

Experimental Immunology, 4, 435-452.

Madureira, J. Ramos, C.I.V., Marques, M., Maia, C., de Sousa, B., Campino, L., M. Santana-

Marques, M.G. and Farrell, N. (2013). Nonclassic metallointercalators with

dipyridophenazine: DNA interaction studies and leishmanicidal activity. Inorganic

Chemistry, 52: 8881-8894.

Malki, A., Laha, R. and Bergmeier, S.C. (2014). Synthesis and cytotoxic activity of MOM-ether

analogs of isosteviol. Bioorganic and Medicinal Chemistry Letters, 24: 1184-1187.

Page 142: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

120

Mandal, G., Mandal, S., Sharma, M., Charret, K.S., Papadopoulou, B., Bhattacharjee, M. and

Mukhopadhyay, R. (2015). Species-specific antimonial sensitivity in Leishmania is

driven by post-transcriptional regulation of AQP1. PLoS Neglected Tropical Diseases, 9

(2): e0003500.

Marcili, A., Sperança, M.A., da Costa, A.P., Madeira, M.F., Soares, H.S., Sanches, C.O.C.C.,

Acosta, I.C.l., Girotto, A., Minervino, A.H.H., Horta M.C., Shaw, J.F. and Gennari,

S.M. (2014). Phylogenetic relationships of Leishmania species based on trypanosomatid

barcode (SSU rDNA) and gGAPDH genes: Taxonomic revision of Leishmania (L.)

infantum chagasi in South America. Infection, Genetics and Evolution, 25: 44-51.

Marques, C.A., Dickens, N.J., Paape, D., Campbell, S.J. and McCulloch, R. (2015). Genome-

wide mapping reveals single-origin chromosome replication in Leishmania, a eukaryotic

microbe. Genome Biology, 16:230: DOI 10.1186/s13059-015-0788-9.

Matsui, M.; Matsui, K.; Kawasaki, Y.; Oda, Y.; Noguchi, T.; Kitagawa, Y.; Sawada1, M.;

Hayashi, M.; Nohmi, T.; Yoshihira, K.; Ishidate Jr, M. and Sofuni, T. (1996).

Evaluation of the genotoxicity of stevioside and steviol using six in vitro and one in vivo

mutagenicity assays. Mutagenesis, 11 (6) 573-579.

Mears, R.E., Modabber, F., Don, R. and Johnson, G.E. (2015). A Review: The current in vivo

models for the discovery and utility of new anti-leishmanial drugs targeting cutaneous

leishmaniasis. PLOS Neglected Tropical Diseases, DOI: 10.1371/journal.pntd.0003889.

Mendes, E.A., Desoti, V.C., Silva, S.deO, Ueda-Nakamura, T., Filho, B.P.D., Yamada-Ogatta,

S.F., Sarragiotto, M.H. and Nakamura, C.V. (2016). C5 induces different cell death

pathways in promastigotes of Leishmania amazonensis. Chemico-Biological

Interactions, 256:16-24.

Mesa-Valle, C.M., Moraleda-Lindez, V., Craciunescu, D. and Osuna, A. (1996). Antileishmanial

Action of Organometallic Complexes of Pt(II) and Rh(I). Memorias Instituto do

Oswaldo Cruz, Rio de Janeiro, 91 (5): 625-633.

Mironov, V.F. and Chestnova, R. V. (2011). Synthesis and antituberculosis activity of

derivatives of Stevia rebaudiana glycoside steviolbioside and diterpenoid isosteviol

containing hydrazone, hydrazide, and pyridinoyl moieties. Russian Journal of

Bioorganic Chemistry, 37 (4):483–491.

Page 143: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

121

Mitropoulos, P., Pete Konidas, P. and Durkin-Konidas, M. (2010). New World cutaneous

leishmaniasis: updated review of current and future diagnosis and treatment. Journal of

Academy of Dermatology, 63 (2): doi: 10.1016/j.jaad.2009.06.088.

Mizushina, Y.; Akihisa, T.; Ukiya, M.; Hamasaki, Y.; Murakami-Nakai, C.; Kuriyama, I.;

Takeuchi, T.; Sugawara, F. and Yoshida, H. (2005). Structural analysis of isosteviol and

related compounds as DNA polymerase and DNA topoisomerase inhibitors. Life

Sciences, 77: 2127–2140.

Momen, H. and Cupolillo, E. (2000). Speculations on the origin and evolution of genus

Leishmania. Memorias Instituto do Oswaldo Cruz, Rio de Janeiro, 95 (04): 583-588.

Momtazi-Borojeni, A.A., Esmaeili, S., Abdollahi, E. and Sahebkar, A. (2016). A Review on the

pharmacology and toxicology of steviol glycosides extracted from Stevia rebaudiana.

Current Pharmaceutical Design, DOI: 10.2174/1381612822666161021142835

Monis, P.T. (1999). The importance of systematic in parasitological research. International

Journal for Parasitology, 29: 381-388.

Monzote, L., Montalvo, A.M., Scull, R., Miranda, M. and Abreu, J. (2007). Activity, toxicity and

analysis of resistance of essential oil from Chenopodium ambrosioides after

intraperitoneal, oral and intralesional administration in BALB/c mice infected with

Leishmania amazonensis: A preliminary study. Biomedicine & Pharmacotherapy, 61:

148-153.

Monzote, L., Pastor, J., Scull, R. and Gille, L. (2014). Antileishmanial activity of essential oil

from Chenopodium ambrosioides and its main components against experimental

cutaneous leishmaniasis in BALB/c mice. Phytomedicine, 21: 1048–1052.

Monzote, L., Stamberg, W., Staniek, S. and Gille, L. (2009). Toxic effects of carvacrol,

caryophyllene oxide, and ascaridole from essential oil of Chenopodium ambrosioides on

mitochondria. Toxicology and Applied Pharmacology, 240: 337–347.

Monzote, L., Montalvo, A.M., Almanonni, S., Scull, R., Miranda, M. and Abreu, J. (2006).

Activity of the essential oil from Chenopodium ambrosioides grown in Cuba against

Leishmania amazonensis. Chemotherapy, 52 (03): 130-136.

Mooney, B.P. (2009). The second green revolution? Production of plant-based biodegradable

plastics. Biochemistry Journal, 418: 219–232.

Page 144: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

122

Moura, M.T., de Sousa, R.V., Leme, L.O. and Rumpf, R. (2008). Analysis of actinomycin D

treated cattle oocytes and their use for somatic cell nuclear transfer. Animal

Reproduction Science, 109: 40–49.

Nagarajan, A. and Brindha, P. (2012). Diterpenes-a review on therapeutic uses with special

emphasis on antidiabetic activity. Journal of Pharmacy Research, 5(8),4530-4540.

Nagle, A.S., Khare, S., Kumar, A.B., Supek, F., Buchynskyy, A., Mathison, C.J.N.,

Chennamaneni, N.K., Pendem, N., Buckner, F.S., Gelb, M.H. and Molteni, V. (2014).

Recent developments in drug discovery for leishmaniasis and human African

Trypanosomiasis. Chemical Reviews, 2014, 114, 11305-11347.

Navarro, M., Cisneros-Fajardo, E.J. and Marchan, E. (2006). New silver polypyridyl complexes:

synthesis, characterization and biological activity on Leishmania mexicana. Drug

Research, 56 (08): 600-604.

Navarro, M., Cisneros-Fajardo, E.J., Fernandez-Mestre, M., Arrieche, D. and Marchan, E.,

(2003). Synthesis, characterization, DNA binding study and biological activity against

Leishmania mexicana of [Cu(dppz)2] BF4. Journal of Inorganic Biochemistry, 97: 364-

369.

Nogueira, M.S., Da Costa, F.B., Brun, R., Kaiser, M. and Schmidt, T.J. (2016). Ent-pimarane

and ent-kaurane diterpenes from Aldama discolor (Asteraceae) and their antiprotozoal

activity. Molecules, 21:1237; doi:10.3390/molecules21091237.

Noosud, J., Lailerd, N., Kayan, A. and Boonkaewwan, C. (2017). In vitro and in vivo assessment

of inhibitory effect of stevioside on pro-inflammatory cytokines. Avicenna Journal of

Phytomedicine, 7 (2): 101-106.

Nowicki, M.W., Tulloch, L.B., Worralll, L., McNae, L.W., Hannaert, V., Michels, P.A.M.,

Fothergill-Gilmore, L.A., Walkinshaw, M.D. and Turner, N.J. (2008). Design, synthesis

and trypanocidal activity of lead compounds based on inhibitors of parasite glycolysis.

Bioorganic and Medicinal Chemistry, 16: 5050-5061.

Nsi-Emvo, E. and Raul, F. (1984). Stimulation of lactase synthesis induced by starvation in the

jejunum of adult rat. Enzyme, 31: 45-49.

Nunes A, Ferreira-Machado S, Nunes R, et al. Analysis of genotoxic potentiality of stevioside by

comet assay. Food and Chemical Toxicology, 45:662-6.

Page 145: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

123

Oliveira, F., Jochim, R.G., Valenzuela, J.G. and Kamhawi, S. (2009). Sand flies, Leishmania,

and transcriptome-borne solutions. Parasitology International, 58: 1–5.

Olivier, M., Gregory, D.J. and Forget, G. (2005). Subversion Mechanisms by which Leishmania

parasites can escape the host immune response: a signaling point of view. Clinical

Microbiology Reviews, 18 (2): 293-205.

Ordóñez-Gutiérrez, L., Espada-Fernández, R., Dea-Ayuela, M.A., Torrado, J.J., Bolas

Fernandez, F. and Alunda, J.M. (2007). In vitro effect of new formulations of

amphotericin B on amastigote and promastigote forms of Leishmania infantum.

International Journal of Antimicrobial Agents, 30: 325–329.

Osipov, A, Arkhangelskaya, E, Vinokurov, A., Smetaninа, N., Zhavoronkov, A. and Dmitry

Klokov, D. (2014). DNA comet giemsa staining for conventional bright-field

microscopy. International Journal of Molecular Science, 15, 6086-6095;

doi:10.3390/ijms15046086

Osipov, A.N., Klokov, D.Y., Elakov, A.L., Rozanova, O.M., Zaichkina, S.I., Aptikaeva, G.F.,

Akhmadieva, A.Kh. (2004). Comparison in vivo study of genotoxic action of high-

versus very low dose-rate γ-irradiation. Nonlinearity in Biology Toxicology and

Medicine, 2: 223–232.

Ostling, O. and Johanson, K.J. (1984). Microelectrophoretic study of radiation-induced DNA

damages in individual mammalian cells. Biochemical and Biophysical Research

Communications, 123, 291–298.

Paila, Y.D., Saha, B. and Chattopadhyay, A. (2010). Amphotericin B inhibits entry of

Leishmania donovani into primary macrophages. Biochemical and Biophysical

Research Communications, 399: 429–433.

Pastor, J., García, M., Steinbauer, S., Setzer, W.N., Scull, R., Gille, L. and Monzote, L. (2015).

Combinations of ascaridole, carvacrol, and caryophyllene oxide against Leishmania.

Acta Tropica, 145: 31–38.

Pathak, M.K., Dhawan, D., Lindner, D.J., Borden, E.C., Farver, C. and Yi, T. (2002).

Pentamidine is an inhibitor of PRL phosphatases with anticancer activity. Molecular

Cancer Therapeutics, 1 (14): 1255-64.

Patrício, F.J., Costa, G.C., Pereira, P.V.S., Aragáo-Filho, W.C., Sousa, S.M., Frazáo, J.B.,

Pereira, W.S., Maciel, M.C.G., Silva, L.A., Amaral, F.M.M., Rebêlo, J.M.M., Guerra,

Page 146: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

124

R.N.M., Ribeiro, M.N.S. and Nascimento, F.R.F. (2008). Efficacy of the intralesional

treatment with Chenopodium ambrosioides in the murine infection by Leishmania

amazonensis. Journal of Ethnopharmacology, 115: 313–319.

Peacock, C.S., Seeger, K., Harris, D., Murphy, L., Ruiz, J.C., Quail, M.A., Peters, N., Adlem, E.,

Tivey, A., Aslett, M., Kerhornou, A., Ivens, A., Fraser, A., Rajandream, M., Carver, T.,

Norbertczak, H., Chillingworth, T., Hance, Z., Jagels, K., Moule, S., Ormond, D.,

Rutter, S., Squares, R., Whitehead, S., Rabbinowitsch, E., Arrowsmith, C., White, B.,

Thurston, S., Bringaud, F., Baldauf, S.L., Faulconbridge, E., Jeffares, D., Depledge,

D.P., Oyola, S.O., Hilley, J.D., Brito, L.O., Tosi, L.R.O., Barrell, B., Cruz, A.K.,

Mottram, J.C., Smith, D.F. and Berriman, M. (2007). Comparative genomic analysis of

three Leishmania species that cause diverse human disease. Nature Genetics, 39 (07):

839–847. doi:10.1038/ng2053.

Pereira, L.L.S., Marcussi, S., Sátiro, L.C., Pereira, C.A., Andrade, L.F., Davide, L.C. and dos

Santo, C.D. (2012) Application of Comet assay to assess the effects of white bean meal

on DNA of human lymphocytes. Brazilian Journal of Pharmaceutical Sciences, 48 (1).

Pérez-Moreno, G., Cantizani, J., SánchezCarrasco, P., Ruiz-Pérez, L.M., Martín, J., el Aouad,

N., Pérez-Victoria, I., Tormo, J.R., González-Menendez, V., González, I., de Pedro, N.,

Reyes, F., Genilloud, O., Vicente, F. and González-Pacanowska, D. (2016). Discovery

of new compounds active against Plasmodium falciparum by high throughput screening

of microbial natural products. PLoS ONE, 11 (1): e0145812.

doi:10.1371/journal.pone.0145812.

Pezzuto, J.M.; Compadre. C.M.; Swanson. S.M.; Nanayakkara, N.P.D. and Kinghorn, A.D.

(1985). Metabolically activated steviol, the aglycone of stevioside, is mutagenic.

Proceedings of National Academy Science, 82, 2478-2482.

Pinto-Martinez, A.K.; Rodriguez-Duran, J.; Serrano-Martin, X.; Hernandez-Rodriguez, V. and

Benaim, G. (2018). Mechanism of action of miltefosine on Leishmania

donovani involves the impairment of acidocalcisome function and the activation of the

sphingosine-dependent plasma membrane Ca2+ channel. Antimicrobial Agents and

Chemotherapy, 62: e01614–e1617.

Ponte-Sucre, A., Gamarro, F., Dujardin, J. C., Barrett, M. P., López-Vélez, R., García-Hernández

R et al. (2017). Drug resistance and treatment failure in leishmaniasis: A 21st century

Page 147: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

125

challenge. PLoS Neglected Tropical Diseases, 11(12): e0006052.

https://doi.org/10.1371/journal.pntd.0006052.

Portas, A.d.S., Miguel, D.C., Yokoyama-Yasunaka, J.K.U. Uliana, S.R.B. and Espósito, B.P.

(2012). Increasing the activity of copper(II) complexes against Leishmania through

lipophilicity and pro-oxidant ability. Journal of Bioinorganic Chemistry, 17: 107-112.

Pujol-Brugués, A., Calpena-Campmany, A.C., Riera-Lizandra, C., Halbaut-Bellowa, L. and

Clares-Naveros, B. (2014). Development of a liquid chromatographic method for the

quantification of paromomycin. Application to in vitro release and ex vivo permeation

studies. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 133:

657–662.

Quigley, G.J., Wang, A. H.J., Ughetto, G., van der Marel, G.A., van Boom, J. H. and Rich, A.

(1980). Molecular structure of an anticancer drug-DNA complex: daunomycin plus d

(CpGpTpApCpG). Proceedings National Academy Science, 77: 7204-7208.

Ramalho-Ortiago, M., Saraiva, E.M. and Traub-Cseko, Y.M. (2010). Sand fly Leishmania

interactions: long relationships are not necessarily easy. The Open Paerasitology

Journal, 4: 195-204.

Reguera, M.R.., Balaña-Fouce R.., Cubría, J.C., Alvarez-Bujidos, M.L. and Ordóñez, D. (1994)

Putrescine uptake inhibition by aromatic diamidines in Leishmania infantum

promastigotes. Biochemical Pharmacology, 47, 1859–1866.

Rehault-Godbert, S., Baron, F., Mignon-Grasteau, S., Labas, V., Gautier, M., Hincke, M.T. and

Nys, Y. (2010). Effect of temperature and time of storage on protein stability and anti-

Salmonella activity of egg white. Journal of Food Protection, 73 (9):1604-12.

Reus, A.A.; Mustafa, U. and Krul, C.A.M. (2012). The use of ex vivo human skin tissue for

genotoxicity testing. Toxicology and Applied Pharmacology, 261 (2): DOI:

10.1016/J.TAAP.2012.03.019.

Ribeiro, T.G., Chávez-Fumagalli, M.A., Valadares, D.G., Franca, J.R., Lage, P.S. Duarte, M.C.,

Andrade, P.H.R., Martins, V.T., Costa, L.E., Arruda, A.L.A., Faraco, A.A.G., Coelho,

E.A.F. and Castilho, R.O. (2014). Antileishmanial activity and cytotoxicity of Brazilian

plants. Experimental Parasitology, 143: 60-68.

Rodrigues, I.A., Mazotto, A.m., Cardoso, V., Alves, R.L., Amaral, A.C.F., Silva, J.R.A.,

Pinheiro, A.S. and Vermelho, A.B. (2015). Natural products: insights into leishmaniasis

Page 148: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

126

inflammatory response. Mediators of Inflammation, Article ID 835910, 12 pages

(http://dx.doi.org/10.1155/2015/835910).

Rodrigues, K.A.daF., Amorim, L.V., Dias, C.N., Moraes, D.F.C., Carneiro, S.M.P. and

Carvalho, F.A.deA. (2015). Syzygium cumini (L.) Skeels essential oil and its major

constituent α-pinene exhibit anti-Leishmania activity through immunomodulation in

vitro. Journal of Ethnopharmacology, 160: 32–40.

Rogers, M., Kropf, P., Choi, B., Dillon, R., Podinovskaia, M., Bates. P. and Müller, I. (2009).

Proteophosophoglycans regurgitated by Leishmania-infected sand flies target the L-

arginine metabolism of host macrophages to promote parasite survival. PLoS

Pathogens, 5: 555-68.

Romano, A., Inbar, E., Debrabant, A., Charmoy, M., Lawyer, P., Ribeiro-Gomes, F., Barhoumi,

M., Grigg, M., Shaik, J., Dobson, D., Beverley, S.M. and Sacks, D.L. (2014). Cross-

species genetic exchange between visceral and cutaneous strains of Leishmania in the

sand fly vector. PNAS, 111 (47): 16808-16813.

Roque, A.L.R. and Jansen, A.M. (2014). Wild and synanthropic reservoirs of Leishmania species

in Americas. International Journal of Parasitology: Parasites and Wildlife, 3: 251-262.

Rowland, M., Munir, A., Durrani, N., Noyes, H. and Reyburn, H. (1999). An outbreak of

cutaneous leishmaniasis in an Afghan Refugees camp in north-west Pakistan.

Transactions of the Royal Society of Tropical Medicine and Hygiene, 93 (2): 133-136.

Rubio J, Calderón JS, Flores A, Castro C, Céspedes CL. 2005. Trypanocidal activity of oleoresin

and terpenoids isolated from Pinus oocarpa. Zeitschrift für Naturforschung, 60: 711–

716.

Sachdeva, H., Sehgal, R. and Kaur, S. (2013). Studies on the protective and immunomodulatory

efficacy of Withania somnifera along with cisplatin against experimental visceral

leishmaniasis. Parasitological Research, 112: 2269–2280.

Sacks, D. L. (1989). Metacyclogenesis in Leishmania promastigotes. Experimental Parasitology,

69:100–103.

Sacks, D. L., and Perkins, P. V. (1984). Identification of an infective stage of Leishmania

promastigotes. Science, 223: 1417–1419.

Page 149: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

127

Saha, A.K., Mukherjee, T. and Bhaduri, M. (1986). Mechanism of action of amphotericin B on

Leishmania dononani promastigotes. Molecular and Biochemical Parasitology, 19:

195-200.

Sakaki, H.; Kakehi, M.; Sadamoto, K.; Nemoto, S. and Kurata, M. (2015). In vitro comet assay

in cultured human corneal epithelial cells. Fundamental Toxicological Sciences, 2 (4):

147-153.

Saraiva, M.E., Pinto-da-Silva, L., Wanderley, J.L.M., Bonomo, A.C., Barcinski, M.A. and

Moreira, M.E.C. (2005). Flow cytometric assessment of Leishmania spp metacyclic

differentiation: Validation by morphological features and specific markers.

Experimental Parasitology, 110: 39-47.

Savoia, D. (2015). Recent updates and perspectives on leishmaniasis. The Journal of Infection in

Developing Countries, 9 (6):588-596.

Schleicher, U., Liese, J., Knippert, I., Kurzmann, C., Hesse, A., Heit, A., Fischer, J.A.A., Weiss,

S., Kalinke, U., Kunz, S. and Bogdan, C. (2008). NK cell activation in visceral

leishmaniasis requires TLR9, myeloid DCs, and IL-12, but is independent of

plasmacytoid DCs. The Journal of Experimental Medicine, 204 (4): 896-906.

Schlein, Y. (1993). Leishmania and sandflies: Interactions in the life cycle and transmission.

Parasitology Today, 9 (7): 255-258.

Schraton, T.M. and Scott, P. (1993). Natural killer cells are the source of interferon γ that drives

differentiation of CD4+ T cell subsets and induces early resistance to Leishmania major

in mice. Journal of Experimental Medicine, 178: 567-577.

Seifert, K. and Croft, S.L. (2006). In Vitro and in vivo interactions between Miltefosine and other

antileishmanial drugs. Antimicrobial Agents and Chemotherapy, 50 (01): 73-79.

Selvapandiyan, A., Dey, R., Gannavaram, S., Solanki, S., Salotra, P. and Nakhasi, H.L. (2014).

Generation of growth arrested Leishmania amastigotes: A tool to develop live

attenuated vaccine candidates against visceral leishmaniasis. Vaccine, 32: 3895–3901.

Sengupta, S. K., Kelly, C. & Sehgal, R. K. (1985) “Reverse” and “symmetrical” analogs of

actinomycin D: metabolic activation and in vitro and in vivo tumor growth inhibitory

activities. Journal of Medicinal Chemistry, 28, 620–628.

Page 150: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

128

Sharma, U., Velpandian, T., Sharma, P. and Singh, S. (2009). Evaluation of anti-leishmanial

activity of selected Indian plants known to have antimicrobial properties.

Parasitological Research, DOI 10.1007/s00436-009-1554-2.

Shivanna, N., Naika, M., Khanum, F. and Kaul, K.K. (2013). Antioxidant, anti-diabetic and renal

protective properties of Stevia rebaudiana. Journal of Diabetes and Its Complications,

27: 103–113.

Siboni, R.B., Nakamori, M., Wagner, S.D., Struck, A.J., Coonrod, L.A., Harriott, S.A., Cass,

D.M., Tanner, M. K. and Berglund, J.A. (2015). Actinomycin D specifically reduces

expanded CUG repeat RNA in myotonic dystrophy models. Cell Reports,13: 2386–

2394.

Silva-Jardim, I., Thiemann, O.H. and Anibal, F.F. (2014). Leishmaniasis and Chagas disease

chemotherapy: a critical review. Journal of Brazilian Chemical Society, 25 (10): 1810-

1823.

Singh, K., Ali, V., Singh, K.P., Gupta, P., Suman, S.S., Ghosh, A.K., Bimal, S., Pandey, K. and

Das, P. (2017). Deciphering the interplay between cysteine synthase and thiol cascade

proteins in modulating Amphotericin B resistance and survival of Leishmania donovani

under oxidative stress. Redox Biology, 12: 350-366.

Singh, N.P., McCoy, M.T., Tice, R.R. and Schneider, E.L. (1988). A simple technique for

quantitation of low levels of DNA damage in individual cells. Experimental Cell

Research, 175: 184–191.

Sinha, A.K., Singh, P., Prakash, A., Pal, D., Dube, K. and Kumar, A. (2017). Putative drug and

vaccine target identification in Leishmania donovani membrane proteins using Naїve

Bayes probabilistic classifier. IEEE/ACM Transactions on Computational Biology and

Bioinformatics, 14 (1): Digital Object Identifier no. 10.1109/TCBB.2016.2570217.

Smanski, M.J., Peterson, R.M. and Shen, B. (2012). Chapter 8 – Platensimycin and platencin

biosynthesis in Streptomyces platnsis, showcasing discovery and characterization of

novel bacterial diterpene synthesis. Methods in Enzymology, 515: 163-186.

Soares, R.P.P., Cardoso, T.L., Barron, T., Araújo, M.S.S., Pimenta, P.F.p. and Turco, S.J. (2005).

Leishmania braziliensis: a novel mechanism in the lipophosphoglycan regulation during

metacyclogenesis. International Journal for Parasitology, 35:245-253.

Page 151: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

129

Sonoki, E., Ng, C.L., Lee, M.C.S., Guo, D., Zhang, Y., Zhou, Y., Alley, M.R.K., Ahyong, V.,

Sanz, L.M., Lafuente-Monasterio, M.J., Dong, C., Schupp, P.G., Gut, J., Legac, J.,

Cooper, R.A., Gamo, F., DeRisi, J., Freund, Y.R., Fidock, D.A. and Rosenthal, P.J.

(2017). A potent antimalarial benzoxaborole targets a Plasmodium falciparum cleavage

and polyadenylation specificity factor homologue. Nature Communications, DOI:

10.1038/ncomms4574.

Soong, L. (2008). Modulation of the dendritic cell function by Leishmania parasites. The Journal

of Immunology, 180: 4355–4360.

Soosaraei, M., Fakhar, M., Teshnizi, S.H., Hezarjaribi, H.Z. and Banimostafavi, E.S. (2017).

Medicinal plants with promising antileishmanial activity in Iran: a systematic review

and meta-analysis. Annals of Medicine and Surgery, 21:63-80.

Soptin, A., Rouhani, S., Ghaemmaghami, P., Haghighi, A., Zolfaghari, M.R., Amirkhani, A.,

Farahmand, A., bordbar, A. and Parvizi, P. (2015). Different morphologies of

Leishmania major amastigotes with no molecular diversity in a neglected endemic area

of zoonotic cutaneous leishmaniasis in Iran. Iranian Biomedical Journal, 19 (03): 149-

159.

Spáth, G.F. and Beverley, S.M. (2001). A lipophosphoglycan-independent method for isolation

of infective Leishmania metacyclic promastigotes by density gradient centrifugation.

Experimental Parasitology, 99: 97–103.

Srivastav, S., Ball, W.B., Gupta, P., Giri, J., Ukil, A. and Das, P.K. (2014). Leishmania donovani

prevents oxidative burst-mediated apoptosis of host macrophages through selective

induction of suppressors of cytokine signaling (socs) proteins. The Journal of Biological

Chemistry, 289 (2):1092–1105.

Sterkers, Y., Crobu, L., Lachaud, L., Pagès, M. and Bastien, P. (2014). Parasexuality and mosaic

aneuploidy in Leishmania: alternative genetics. Trends in Parasitology, 30 (09): 429-

435.

Steverding, D. (2017). The history of Leishmaniasis. Parasite & Vectors, 10 (1):82

doi: 10.1186/s13071-017-2028-5

Strobykina, I.Y., Belenok, M.G., Semenova, M.N., Semenov, V.V., Babaev, V.M., Rizvanov,

I.K., Mironov, V.F. and Kataev, V.E. (2015). Triphenylphosphonium cations of the

Page 152: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

130

diterpenoid isosteviol: synthesis and antimitotic activity in a sea urchin embryo model.

Journal of Natural Products, 78 (6): 1300-8. DOI: 10.1021/acs.jnatprod.5b00124

Suankratay, C., Suwanpimolkul, G., Wilde, H. and Siriyasatien, P. (2010). Case report:

Autochthonous visceral leishmaniasis in a Human Immunodeficiency Virus (HIV)-

infected patient: the first in thailand and review of the literature. American Journal of

Tropical Medicine and Hygiene, 82 (1): 4-8.

Sundar, S. and Singh, A. (2016). Recent developments and prospects in the treatment of visceral

leishmaniasis. Therapeutic Advances in Infectious Disease, 3 (3-4): 98-109.

Sunyoto, T., Potet J and Boelaert, M. (2018). Why miltefosine-a life-saving drug for

leishmaniasis-is unavailable to people who need it the most. BMJ Global Health, 2018;

3: e000709. Doi: 10.1136/ bmjgh-2018-000709.

Suttajit, M.; Vinitketkaumnuen, U.; Meevatee, U.; Buddhasukh, D. (1993). Mutagenicity and

human chromosomal effect of stevioside, a sweetener from Stevia rebaudiana Bertoni.

Environ. Health Perspective, 101 (Suppl.), 53–56.

Tahghighi, A. (2014). Importance of metal complexes for development of potential

leishmanicidal agents. Journal of Organometallic Chemistry, 770: 51-60.

Takasaki, M., Konoshima, T., Kozuka, M., Tokuda, H., Takayasu, J., Nishino, H., Miyakoshi,

M., Mizutani, K. and Lee, K. (2009) Cancer preventive agents. Part 8: Chemo

preventive effects of stevioside and related compounds. Bioorganic & Medicinal

Chemistry, 17: 600–605.

Teixeira, M. C. A. (2002). "A simple and reproducible method to obtain large numbers of axenic

amastigotes of different Leishmania species. Parasitological Research, 88: 963-968.

Theophilus, P. A. S., Victoria, M. J., Socarras, K. M., Filush, K. R., Gupta, K., Luecke, D. F. and

Sapi, E. (2015). Effectiveness of Stevia rebaudiana whole leaf extract against the

various morphological forms of Borrelia burgdorferi in vitro. European Journal of

Microbiology and Immunology, 5 (4): 268–280.

Tice, R.R., Agurell, E., Anderson, D., Burlinson, B., Hartmann, A., Kobayashi, H., Iyamae, Y.,

Rojas, E., Ryu, J.C., Sasaki, Y.F. (2000). Single cell gel/comet assay: guidelines for in

vitro and in vivo genetic toxicology testing. Environmental and Molecular Mutagen, 35

(3): 206-221.

Page 153: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

131

Toumatia, O., Yekkour, A., Goudjal, Y., Riba, A., Coppel, Y., Mathieu, F., Sabaou, N. and

Zitouni, A. (2015). Antifungal properties of an actinomycin D-producing strain,

Streptomyces sp. IA1, isolated from a Saharan soil. Journal Basic Microbiology, 2015,

55, 221–228.

Tripathi, C.D.P., Gupta, R., Kushawaha, P.K., Mandal, C., Bhattacharya, S.M. and Dube, A.

(2014). Efficacy of Withania somnifera chemotypes NMITLI – 101R, 118R and

withaferin A against experimental visceral leishmaniasis. Parasite Immunology, 36:

253–265.

Tripathi, C.D.P., Kushawaha, P.K., Sangwan, R.S., Mandal, C., Misra-Bhattacharya, S. and

Dube, A. (2017). Withania somnifera chemotype NMITLI 101R significantly increases

the efficacy of antileishmanial drugs by generating strong IFN- γand IL-12 mediated

immune responses in Leishmania donovani infected hamsters. Phytomedicine, 24: 87–

95.

Tuon, F.F., Amato, V. and Amato, N.V. (2008). Leishmania: origin, evolution and future since

the Precambrian. FEMS Immunology and Medical Microbiology, 54: 158-166.

Ullah, A., Baratto, L.C., Paula, R.C., Silva, L.H.V., Soares, M.J. and Oliveira, B.H. (2016).

Preparation of derivatives of betulinic acid, steviol and isosteviol and evaluation

of antitrypanosomal and antimalarial activities. Journal of Brazilian Chemical Society,

27 (7): 1245-1253.

Ullah, A., Munir, S., Makhboot, Y. and Badsha, S.L. (2019). Bioactivity profile of diterpenoid

Isosteviol and its derivatives. Molecules, 24: 678. DOI: 10.3390/molecules24040678.

Valeriote, F., Vietti, T. and Tolen, S. (1973). Kinetics of the lethal effect of actinomycin D on

normal and leukemic cells. Cancer Research, 33: 2658-2661.

Vannier-Santos, M.A., Martiny, A. and de Souza, W. (2002). Cell biology of Leishmania spp.:

invading and evading. Current Pharmaceutical Design, 8: 297-318.

Varela-M, R.E., Villa-Pulgarin, J.A., Yepes, E., Müller, I., Modolell, M., Munõz, D.L., Robledo,

S.M., Muskus, C.E., López-Abán, J., Muro, A., Vélez, I.D. and Mollinedo, F. (2012). In

vitro and in vivo efficacy of ether lipid edelfosine against Leishmania spp. and SbV-

resistant parasites. PLoS Neglected Tropical Diseases, 6 (4): e1612.

doi:10.1371/journal.pntd.0001612.

Page 154: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

132

Volpe, D.A., Hamed, S.S. and Zhang, L.K. (2014). Use of different parameters and equations for

calculation of IC50 values in efflux assays: potential sources of variability in IC50

determination. The AAPS Journal, 16 (1): DOI: 10.1208/s12248-013-9554-7.

Vorobyeva, N.Y., Osipov, A.N. and Pelevina, I.I. (2007). Sensitivity of peripheral blood

lymphocytes of pilots and astronauts to gamma-radiation: Induction of double-stranded

DNA breaks. Bulletin of Experimental biology and Medicine, 144; 523–526.

Walker, N.I., Harmon, B.V., Gobe, G.C and Kerr, J.F. (1988). Patterns of cell death. Methods

Achieven in Experimental Pathology, 13, 18–54.

Wang, M., Li, H., Xu, F., Gao, X., Li, J., Xu, S., Zhang, D., Wu, X., Xu, J., Hua, H. and Li, D.

(2018). Diterpenoid lead stevioside and its hydrolysis products steviol and isosteviol:

biologiclal activity and structural modification. European Journal of Medicinal

Chemistry, 156: 885-906.

Waseem, D., Butt, A.F., Haq, I., Bhatti, M.H. and Khan, G.M. (2017). Carboxylate derivatives of

tributyltin (IV) complexes as anticancer and antileishmanial agents. DARU Journal of

Pharmaceutical Sciences, 25 (08): DOI 10.1186/s40199-017-0174-0

Wheeler, J.R., Gluenz, E. and Gull, K. (2011). The cell cycle of Leishmania: morphogenetic

events and their implications for parasite biology. Molecular Microbiology, 79 (3), 647–

662.

WHO (2016). Giemsa stain of malaria blood films. MM-SOP-07A.

WHO (2019). Leishmaniasis. https://www.who.int/news-room/fact-sheets/details/leishmaniasis.

Yadav, A., Amit, A., Chaudhary, R., Chandel, A.S., Mahantesh, V., Suman, S.S., Singh, S.K.,

Dikhit, M.R., Ali, V., Rabidas, V., Pandey, K., Kumar, A., Das, P. and Bimal, S. (2015).

Leishmania donovani: impairment of the cellular immune response against recombinant

ornithine decarboxylase protein as a possible evasion strategy of Leishmania in visceral

leishmaniasis. International Journal for Parasitology, 45: 33–42.

Yousuf, M., Mukherjee, D., Dey, S., Pal, C. and Adhikari, S. (2016). Antileishmanial

ferrocenylquinoline derivatives: Synthesis and biological evaluation against Leishmania

donovani. Europen Journal of Medicinal Chemistry, 124: 468-479.

Zamora-Chimal, J., Fernández-Figueroa, E.A., Ruiz-Remigio, A., Wilkins-Rodríguez, A.A.,

Delgado-Domínguez, J., alaiza-Suazo, N., Gutiérrez-Kobeh, L. and Becker. (2017).

Page 155: ANTILEISHMANIAL, CYTOTOXIC AND GENOTOXIC EFFECTS OF

133

NKT cell activation by Leishmania mexicana LPG: Description of a novel pathway.

Immunobiology, 222: 454-462.

Zamora-Chimal, J., Hernández-Ruiz, J. and Becker, I. (2016). NKT cells in leishmaniasis.

Immunobiology. http://dx.doi.org/10.1016/j.imbio.2016.11.014.

Zauli-Nascimento, R.C., Miguel, D.C., Yokoyama-Yasunaka, J.K.U., Pereira, L.I.A., de

Oliveira, M.A.P., Rebier0-Dias, F., Dorta, M.L. and Uliana, S.R.B. (2010). In vitro

sensitivity of Leishmania (Viannia) braziliensis and Leishmania (Leishmania)

amazonensis Brazilian isolates to meglumine antimoniate and amphotericin B. Tropical

Medicine and International Health, 15 (1): 68-76.

Zaverucha do Valle, T., Calabrese, K.S., Côrte-Real, S., Baetas, W.C. and Gonçalves da Cota,

S.C. (2003). Trypanosoma cruzi: In vitro morphological alterations induced by

actinomycin D. Pharmacology, 67:55–58.

Zhang, Q., Yang, H., Li, Y., Liu, H. and Jia, X. (2017). Toxicological evaluation of ethanolic

extract from Stevia rebaudiana Bertoni leaves: Genotoxicity and subchronic oral

toxicity. Regulatory Toxicology and Pharmacology, 86 (2017) 253-259.

Zhu, S., Wu, Y., Liu, C., Wei, C., Tao, J. and Liu, H. (2013). Design and stereoselective

synthesis of novel isosteviol-fused pyrazolines and pyrazoles as potential anticancer

agents. European Journal of Medicinal Chemistry, 65 (2013) 70-82.

Zotchev, S.B. (2012). Marine actinomycetes as an emerging resource for the drug development

pipelines. Journal of Biotechnology, 158: 168–175.