universiti putra malaysia therapeutic potentials of …psasir.upm.edu.my/id/eprint/76361/1/fpsk(p)...

76
UNIVERSITI PUTRA MALAYSIA THERAPEUTIC POTENTIALS OF BONE MARROW DERIVED MESENCHYMAL STEM CELLS IN AVERTING ORGAN DAMAGE DUE TO RIFAMPICIN INDUCED TOXICITY IN ANIMAL MODEL DANJUMA LAWAL FPSK(P) 2018 38

Upload: others

Post on 29-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

UNIVERSITI PUTRA MALAYSIA

THERAPEUTIC POTENTIALS OF BONE MARROW DERIVED

MESENCHYMAL STEM CELLS IN AVERTING ORGAN DAMAGE DUE TO RIFAMPICIN INDUCED TOXICITY IN ANIMAL MODEL

DANJUMA LAWAL

FPSK(P) 2018 38

© COPYRIG

HT UPM

i

THERAPEUTIC POTENTIALS OF BONE MARROW DERIVED

MESENCHYMAL STEM CELLS IN AVERTING ORGAN DAMAGE DUE

TO RIFAMPICIN INDUCED TOXICITY IN ANIMAL MODEL

By

DANJUMA LAWAL

Thesis Submitted to the School of Graduate Studies, Universiti Putra Malaysia,

in Fulfillment of the Requirements for the Degree Doctor of Philosophy

Febuary 2018

© COPYRIG

HT UPM

ii

COPYRIGHT

All material contained within the thesis, including without limitation text, logos,

icons, photographs, and all other artwork, is copyright material of Universiti Putra

Malaysia unless otherwise stated. Use may be made of any material contained within

the thesis for non-commercial purposes from the copyright holder. Commercial use

of material may only be made with the express, prior, written permission of

Universiti Putra Malaysia.

Copyright © Universiti Putra Malaysia

© COPYRIG

HT UPM

iii

DEDICATIONS

To the memory of my beloved father may his soul rest in peace, and my beloved

mother both of them have been everything to me.

© COPYRIG

HT UPM

i

Abstract of thesis presented to the Senate of Universiti Putra Malaysia in fulfilment

of the requirement for the degree of Doctor of Philosophy

THERAPEUTIC POTENTIALS OF BONE MARROW DERIVED

MESENCHYMAL STEM CELLS IN AVERTING ORGAN DAMAGE DUE

TO RIFAMPICIN INDUCED TOXICITY IN ANIMAL MODEL

By

DANJUMA LAWAL

Febuary 2018

Chairman : Suresh Kumar Subbiah, PhD

Faculty : Medicine and Health Sciences

Being the front line medicine against tuberculosis, rifampicin has been used for quite

a long period of time. Although it is still effective in killing the bacteria, the drug has

been shown to be associated with many adverse effects, like liver and kidney

toxicity. Investigations of toxicological effects due to rifampicin treatment have been

published since 1974. Just few years after the drug became available in the market

and more and more articles are still published showing its adverse effects on liver

and kidney. Yet the drug is still prescribed and considered the best option. Prolong

rifampicin therapy due to tuberculosis and the toxicity of the drug; coupled with the

proportionate risk of kidney/liver malfunction has stressed the need for a new

interventional approach. Intravenous administration of bone marrow mesenchymal

stem cells along with rifampicin can yield a promising result. This is because the

hepatocytes and renal cells growth factors that are known to have multiple function

like stimulating antiapoptotic and antioxidant actions that can neutralized the

toxicological impacts of the drug can be released by the MSCs. Also the ability of

the transplanted MSCs to differentiate into liver and kidney cells can help in the

organ regeneration as well. This research was design to assess the therapeutic

potential of MSCs in averting organ damage due to rifampicin-induced liver and

kidney toxicity on wistar rats and their progeny. Both male and female wistar rats

were given the therapeutic doses of rifampicin via oral gavage (9mg/kg/day for

3month), rifampicin plus MSCs infusion intravenously 2.5x105cells (twice/month for

3-months), and a control group received normal saline only via oral gavage.

Alteration in biochemical indicators like ALT, AST, total bilirubin, albumin, total

cholesterol, triglycerides, LDL-cholesterol level, HDL-cholesterol level, urea,

creatinine, total protein were found. Pathological changes in both liver and kidney of

the rifampicin treated rats like necrosis of hepatocytes, cytoplasmic vacuolation,

distended sinusoids, loss of polyhedral structure in the liver, hypertrophied of kupper

© COPYRIG

HT UPM

ii

cells in sinusoids, degeneration of liver cells, hypertrophy of hepatocytes, with

pycnotic nuclei and deformed nuclei, disorganization of hepatocytes with lysis of

cytoplasm. Pathological changes in kidney like increase in size of glomeruli and

degeneration of renal tubules were also noticed. In order to determine if these effects

can be transferred to their progeny, both the males and females were breed (while the

treatments continue during the breeding) and the biochemical markers and the

histopathological damages in the progeny were assessed. Transplanted MSCs was

able to avert these effects by the release of growth factors and also by the

differentiation potentials of the cells to both the liver and kidney cells. Transplanted

MSCs showed a promising hepatic and renal protection against rifampicin induced

toxicity in wistar rats and their progeny; as seen in both the biochemistry and

histological data, therefore it can be used in conjunction with the antituberculosis

drug rifampicin. Furthermore, gene expression studies revealed some genes that

were either up regulated or down regulated due to the adverse effects of the drug, but

were gradually returning back to their normal fold change value as a result of the

stem cell treatment. It can be concluded that administration of bone marrow-derived

mesenchymal stem cells have shown some modulatory, regenerative and therapeutic

activity against liver and kidney injury due to prolonged rifampicin treatment in

Wistar rats and their progenies as seen both in the biochemical indicators,

histological assessment, cell quantification and also the gene expression studies.

© COPYRIG

HT UPM

iii

Abstrak tesis yang dikemukakan kepada Senat Universiti Putra Malaysia sebagai

memenuhi keperluan untuk ijazah Doktor Falsafah

POTENSI TERAPEUTIK SEL STEM MESENKIMAL DARI SUM-SUM

TULANG DALAM MENGHINDARI KEROSAKAN ORGAN AKIBAT DARI

KETOKSIKAN DIARUH OLEH RIFAMPISIN DALAM MODEL HAIWAN

Oleh

DANJUMA LAWAL

Februari 2018

Pengerusi : Suresh Kumar Subbiah, PhD

Fakulti : Perubatan dan Sains Kesihatan

Sebagai ubat rawatan utama bagi jangkitan tuberkulosis, rifampisin telah lama

digunakan. Walaupun masih efektif dalam membunuh bakteria ini, rifampisin

dikaitkan dengan pelbagai kesan sampingan seperti kesan toksik pada hati dan buah

pinggang. Penyelidikan mengenai kesan tosikologi penggunaan rifampisin telah

diterbitkan semenjak tahun 1974. Beberapa tahun selepas ubatan ini berada di

pasaran dan semakin banyak artikel yang diterbitkan telah membincangkan

mengenai kesan sampingannya. Walau bagaimanapun, ubat ini masih digunakan dan

masih lagi di ertimbangkan sebagai pilihan yang terbaik untuk rawatan tuberkulosis.

Pengambilan terapi rifampisin dalam jangka masa yang lama dan tahap toksik ubatan

ini, ditambah dengan kesan kepada kegagalan fungsi buah pinggang/hati telah

menyebabkan satu kaedah lain diperlukan untuk pendekatan intervensi baru.

Penyaluran sel stem mesenkimal (MSCs) dari sum-sum tulang secara interavenus

bersama dengan rifampisin mampu memberikan keputusan yang

memberangsangkan. Oleh kerana sel hepatosit dan faktor penggalak sel renal telah

diketahui mempunyai pelbagai fungsi dalam menggalakan kesan anti-apoptosis dan

antioksidan dalam meneuteralkan impak toksikologikal ubatan ini dan kebolehan

MSCs untuk berubah kepada sel hati dan sel buah pinggang, kedua-dua fenomena ini

dijangka dapat membantu dalam pembaikian organ tersebut. Penyelidikan ini

dibentuk bertujuan untuk mengetahui potensi terapeutik penggunaan MSCs dalam

pengurangan kesan kerosakan organ akibat dari penggunaan rifampisin yang

menyumbang kepada toksiksiti kepada hati dan buah pinggang bagi tikus wistar dan

keturunannya, Tikus wistar jantan dan betina telah diberikan dos terapeutik

rifampisin 9mg/kg/hari untuk 4 bulan, rifampisin dan MSCs 2 kali/sebulan untuk

tempoh 3 bulan infusi secara intravena serta kumpulan kawalan yang hanya

menerima larutan salin sahaja menerusi oral ‘gavage’. Perubahan dalam indikator

biokimia seperti ALT, AST, jumlah bilirubin, albumin, jumlah kolesterol, trigliserid,

© COPYRIG

HT UPM

iv

paras kolesterol LDL, paras kolesterol HDL, urea, kreatinin, jumlah protein juga

telah direkodkan serta perubahan patologikal dalam hati dan buah pinggang juga

telah dikesan dalam tikus yang diberikan rifampisin seperti nekrosis pada hepatosit,

vakuol sitoplasmik, pengembangan sinusoid, perubahan struktur polihedral pada sel

hati, pembesaran pada sel kupper dalam sinusoid, penguraian sel hati, hipertropi

pada hepatosit serta nuklei yang ‘pycnotik’ dan nuklei yang tidak sempurna,

hepatosit yang tidak teratur dengan sitoplasma yang telah pecah. Buah pinggang juga

mengalami perubahan patologikal seperti penambahan saiz glomeruli serta

penguraian tubul renal juga telah diambil perhatian. Untuk meneliti adakah kesan ini

diwarisi kepada keturunan tikus ini, kedua-dua tikus jantan dan betina telah

dikacukkan dan penanda biokimia dan kerosakan histopatologikal kepada keturunan

telah disiasat. MSCs yang di transplan telah berjaya membendung kesan buruk

ubatan ini dengan penghasilan faktor pembesaran dan juga potensi pembiakan sel

bagi kedua-dua sel hati dan buah pinggang. MSCs yang di transplan menunjukkan

perlindungan yang memberangsangkan kepada heptik dan renal terhadap rifampisin

yang di rangsangkan dalam tikus wistar serta keturunannya, seperti yang di

tunjukkan dari segi biokimia dan data histologikal. Oleh itu, rawatan ini boleh di

gunakan bersama dengan ubatan anti-tuberkulosis, rifampisin. Di sampling itu,

penyelidikan ekspresi gen juga menunjukkan gen terbabit samaada ‘upregulated’

mahupun ‘downregulated’ berpunca dari kesan pengambilan ubat ini. Walau

bagaimanapuan, ianya akan kembali kepada perubahan lipatan yang normal kesan

dari rawatan sel stem. Hal ini menunjukkan bahawa pengambilan sel stem

mesenkima diperolehi daripada sum-sum tulang menunjukkan aktiviti modulasi,

penjanaan semula serta aktiviti terapeutik terhadap kerosakkan pada organ hati dan

buah pinggang akibat rawatan menggunakan rifampisin dalam tempoh masa yang

lama dalam tikus wistar serta progeninya dapat dilihat kesannya dari segi penanda

aras biokimia, penilaian, histologi, penjumlahan sel serta kajian ekspresi gen.

© COPYRIG

HT UPM

v

ACKNOWLEDGEMENTS

All praises are for Almighty ALLAH, the most Beneficent, ever Merciful, countless

thanks to Him, Lord of lords who guides us in darkness and helps in difficulties. And

all respects and regards to Holy prophet (peace be upon him) for enlightening our

conscience with the essence of faith in Allah, converging all his kindness and mercy

upon us. It is a great pleasure and honour for me to express my profound and cordial

gratitude to Dr. Suresh Kumar for his learned guidance, skilled advice and

sympathetic attitudes during the course of the work. All words in my knowledge just

fall to pay tribute to Dr. Mok Pooi ling, for her marvelous and splendid guidance,

winsome attitude, rewardless efforts and nice co-operation in the completion of the

work, I am grateful to Dr. Rukman Awang Hamat. I am also thankful to my team

members Sakinah Bt Maideensa Syed Gulam Rasul, Hiba, Sharmilah Kumari, Amira

and Poorani and to postgraduate students of medical microbiology and stem cell

research lab for their supports. I am also thankful to the technologists in the

Department of Medical Microbiology and Parasitology, Stem cell Research lab and

Haematology lab for their support during the work. I am also thankful to Federal

University Dutse for giving me the opportunity to pursue this programme. Lastly I

am also thankful to my beloved Mother (Maimunatu), my wife (Salma), my children

Maimunatu (Mama), Hafsat (Ummi), Mohammad Auwal (Abba), brothers and

sisters and friends at Department of Microbiology and Biotechnology, Federal

University Duste, for their encouragement during the research journey, May the God

Almighty reward you abundantly.

© COPYRIG

HT UPM

© COPYRIG

HT UPM

vii

This thesis was submitted to the Senate of the Universiti Putra Malaysia and has

been accepted as fulfilment of the requirement for the degree of Doctor of

Philosophy. The members of the Supervisory Committee were as follows:

Suresh Kumar Subbiah, PhD

Senior Lecturer

Faculty of Medicine and Health Sciences

Universiti Putra Malaysia

Chairperson

Mok Pooi Ling, PhD

Senior Lecturer

Faculty of Medicine and Health Sciences

Universiti Putra Malaysia

Member

Rukman Awang Hamat, PhD

Associate Professor

Faculty of Medicine and Health Sciences

Universiti Putra Malaysia

Member

ROBIAH BINTI YUNUS, PhD

Professor and Dean

School of Graduate Studies

Universiti Putra Malaysia

Date:

© COPYRIG

HT UPM

viii

Declaration by graduate student

I hereby confirm that:

• this thesis is my original work;

• quotations, illustrations and citations have been duly referenced;

• this thesis has not been submitted previously or concurrently for any other degree

at any institutions;

• intellectual property from the thesis and copyright of thesis are fully-owned by

Universiti Putra Malaysia, as according to the Universiti Putra Malaysia

(Research) Rules 2012;

• written permission must be obtained from supervisor and the office of Deputy

Vice-Chancellor (Research and innovation) before thesis is published (in the

form of written, printed or in electronic form) including books, journals,

modules, proceedings, popular writings, seminar papers, manuscripts, posters,

reports, lecture notes, learning modules or any other materials as stated in the

Universiti Putra Malaysia (Research) Rules 2012;

• there is no plagiarism or data falsification/fabrication in the thesis, and scholarly

integrity is upheld as according to the Universiti Putra Malaysia (Graduate

Studies) Rules 2003 (Revision 2012-2013) and the Universiti Putra Malaysia

(Research) Rules 2012. The thesis has undergone plagiarism detection software

Signature: Date:

Name and Matric No: Lawal Danjuma (GS43217)

© COPYRIG

HT UPM

ix

Declaration by Members of Supervisory Committee

This is to confirm that:

• the research conducted and the writing of this thesis was under our

supervision;

• supervision responsibilities as stated in the Universiti Putra Malaysia

(Graduate Studies) Rules 2003 (Revision 2012-2013) were adhered to.

Signature:

Name of Chairman

of Supervisory

Committee:

Dr. Suresh Kumar Subbiah

Signature:

Name of Member

of Supervisory

Committee:

Dr. Mok pooi Ling

Signature:

Name of Member

of Supervisory

Committee:

Dr. Rukman Awang Hamat

© COPYRIG

HT UPM

x

TABLE OF CONTENTS

Page

ABSTRACT i

ABSTRAK iii

ACKNOWLEDGEMENTS v

APPROVAL vi

DECLARATION viii

LIST OF TABLES xv

LIST OF FIGURES xviii

LIST OF ABBREVIATIONS xxi

CHAPTER

1 INTRODUCTION 1 1.1 Background 1 1.2 Statement of the Problem 4 1.3 Justification for the Study 4

1.4 Hypothesis 4 1.5 General Objective 4 1.6 Research outlook 5

2 LITERATURE REVIEW 10

2.1 Tuberculosis 10 2.2 Drug Impacts on Liver 11

2.2.1 Structure and function of the liver 11 2.2.2 Overall views due to drug-induced liver injury 11

2.2.3 Extents of the Predicament 12 2.2.4 Origin and development of DILI 12 2.2.5 Liver Function Enzyme Quantification 12

2.2.6 Categories of DILI 13 2.2.7 Hepatic acclimatization 13

2.2.8 Drug-induced acute hepatitis or hepatocellular injury 13 2.2.9 Nonalcoholic fatty liver disease 13 2.2.10 Hepatitis due to granuloma 14

2.2.11 Cholestasis 14 2.3 DILI During Medication of Tatent TB Infection 14

2.3.1 Rifampicin 14

2.3.2 Processes of liver toxicity 15

2.3.3 Drug interfaces 15 2.3.4 Medical features of liver toxicity 15 2.3.5 General liver toxicity 16

2.4 Drug Impacts on Kidney 16 2.4.1 Kidney 16

2.4.2 Pathogenic Processes 17 2.4.2.1 Altered IntraGlomerular Hemodynamics 17

© COPYRIG

HT UPM

xi

2.4.2.2 Tubular Cell Toxicity 17

2.4.2.3 Inflammation 17 2.4.2.4 Crystal Nephropathy 18 2.4.2.5 Rhabdomyolysis 19

2.4.2.6 Thrombotic Microangiopathy 19 2.4.3 Rifampicin 19

2.5 Stem Cells and Infectious Disease 20 2.5.1 Variation in Expression of Surface Marker Antibodies

on Bone MarrowMesenchymal Stem Cell in Rats and Mice

as Compared with Human 21 2.5.2 Three major criteria of MSCs 25 2.5.3 Therapeutic Potential of MSCs 25

2.5.3.1 Competency to Move and Graft 26 2.5.3.2 Delineation or Differentiation 26

2.5.3.3 Production of Several Bioactive Particles 27 2.5.3.4 Immunomodulatory Roles of MSCs 28

2.6 Bone Marrow Derived Mesenchymal Stem Cells and their roles

in organ Repair 29 2.6.1 BMMSCs and its Role in Kidney Treatment 29 2.6.2 BMMSCs and its Role in Liver Treatment of Diseases 29

2.7 Clinical Usage of Bone marrow Derived Stem cells 30 2.8 MSCs for liver Disorder 31

2.8.1 Liver redevelopment or regeneration 31

2.8.2 Cellular processes associated with liver redevelopment 32 2.8.3 Cell based medication for liver redevelopment 33

2.8.4 Concise Clinical Tests with BMMSCs in human with

Liver Disorder 33

2.9 MSCs for the Treatement of kidney Disorders 35 2.9.1 Acute Kidney Disorder 35

2.9.2 Kidney Replacement or Transplantation 37 2.9.3 Chronic Kidney Disorder 38 2.9.4 Preclinical Trials of Chronic Kidney Disorder Treatment

using BMSCs 40 2.9.5 Clinical Tests Involing Mesenchymal Stem Cells for

Kidney Restoration 41 2.9.6 Paracrine Processes of BMSCs Treatment in Acute

renal failure (ARF) 42

2.10 Appropriate Animal Prototype for Tuberculosis Investigation 43 2.11 Molecular Investigations of Liver Damage due to Anti-

tuberculosis Drug Treatment 44

2.12 Molecular Investigation of Kidney Damage due to Anti-

tuberculosis Drug Treatment 47

3 ISOLATION, EXPANSION AND CHARACTERIZATION OF

BONE MARROW DERIVED MESENCHYMAL STEM CELLS

FROM WISTAR RAT 48

3.1 Introduction 48 3.2 Materials and method 48

© COPYRIG

HT UPM

xii

3.2.1 Isolation of Bone Marrow Derived Mesenchymal Stem

cells from wistar Rat 48 3.2.2 Procedure 48

3.3 Expansion and Passaging of bone marrow derived mesenchymal

stem cells 49 3.3.1 Procedure 49

3.4 Cell Counting 50 3.4.1 Preparing hemocytometer 50 3.4.2 Preparing cell suspension 50

3.4.3 Counting 50 3.5 Cryopreservation of Mesenchymal Stem Cells 51

3.5.1 Media and Solutions 51 3.5.2 Procedure 51

3.6 Thawing of Cryopreserved Cells 51

3.7 Immunophenotyping Characterization 52 3.8 Tri-lineage Differentiation and staining 52

3.8.1 Preparation of Reagents 52 3.8.2 Preparation of Osteogenesis Induction Medium: 53 3.8.3 Preparation of Mesenchymal Stem Cell Expansion Medium 53 3.8.4 Osteogenesis Differentiation (for 24-well tissue culture

plates) 54 3.8.5 Cell Plating 54 3.8.6 Alizarin Red Staining Protocol 54

3.8.7 Preparation of Reagents 54 3.8.8 Preparation of Adipogenesis Induction Medium: 55

3.8.9 Preparation of Adipogenesis Maintainace Medium: 55 3.8.10 Preparation of Mesenchymal Stem Cell Expansion Medium 56

3.8.11 Adipogenesis Differentiation 56 3.8.12 Oil red O staining protocol: 57

3.8.13 Preparation of Chondrogenic Differentiation (CD) Medium 57 3.8.14 Chondrogenesis differentiation 58 3.8.15 Alcian blue staining 58

3.9 Results 58 3.9.1 Isolation, expansion and characterization of bone

marrow derived mesenchymal stem cells from wistar rat 58 3.9.2 Isolated bone marrow derived mesenchymal stem cells

from wistar rat 59

3.9.3 Immunophenotyping characterization of bone marrow

derived mesenchymal stem cells from wistar rat 60

3.9.4 Osteoblast detection in bone marrow derived

mesenchymal stem cells 61

3.9.5 Adipocyte detection in bone marrow derived

mesenchymal stem cell 61 3.9.6 Chondrogenic detection in bone marrow

derived mesenchymal stem cell 62 3.10 Discussion 63

3.11 Conclusion 63

© COPYRIG

HT UPM

xiii

4 EFFECTIVENESS OF TRANSPLANTED BONE MARROW

DERIVED MESENCHYMAL STEM CELLS IN AVERTING

ORGAN DAMAGE DUE TO RIFAMPICIN TREATMENT 65 4.1 Introduction 65

4.2 Materials and method 65 4.2.1 Sample Size Calculation for Animal Studies 65 4.2.2 Grouping and Treatment of Animal 66 4.2.3 Administration of Rifampicin 67 4.2.4 MSCs infusion (Administration of MSCs) 67

4.2.5 Collection of serum 67 4.2.6 Organ extraction 68 4.2.7 Histological Analysis 68 4.2.8 Statistical Analysis 68

4.3 Results 68

4.3.1 Blood biochemistry Parameters of Rat and their progeny

after 4-month treatment with rifampicin and rifampicin

plus bone marrow derived mesenchymal stem cells 68 4.3.2 Histological examination of kidney tissue from the parent

rats 81 4.3.3 Histological examination of kidney tissue from progeny

rats 84 4.3.4 Histological examination of liver tissue from the parent rats 86 4.3.5 Histological examination of liver tissue from progeny rats 88

4.4 Discussion 96 4.5 Conclusion 102

5 EFFECTIVENESS OF TRANSPLANTED BONE MARROW

DERIVED MESENCHYMAL STEM CELLS IN CORRECTING

DYSREGULATED GENES DUE TO RIFAMPICIN

TREATMENT 103 5.1 Introduction 103 5.2 Materials and method 103

5.2.1 Total RNA Extraction and RNA integrity number

(RIN) Assessment 103

5.2.2 Library Preparation and Quality Checking (QC) 104 5.2.3 mRNA Isolation, Fragmentation and Priming Starting

with Total RNA 104

5.2.4 Preparation of First Strand Reaction Buffer and

Random Primer Mix 105

5.2.5 First Strand cDNA Synthesis 105

5.2.6 Second Strand cDNA Synthesis 106

5.2.7 Purifying the Double-stranded cDNA Using 1.8X

Agencourt AMPure XP Beads 106 5.2.8 End Prep of cDNA Library 107 5.2.9 Adaptor Ligation 107 5.2.10 Purifying the Ligation Reaction Using AMPure XP Beads 108

5.2.11 PCR Library Enrichment 109 5.2.12 Purification of the PCR Reaction using Agencourt

© COPYRIG

HT UPM

xiv

AMPure XP Beads 109

5.2.13 Assessing library quantity and quality on Qubit

and Bioanalyzer (Qubit Fluorometer 2.0 & Agilent 2100

High Sensitivity Chip) 110

5.3 Library Sequencing 110 5.4 Gene Expression Analysis (Transcriptomic construction or

Read mapping & quantification) 110 5.5 Results 111

5.5.1 Mapping and Annotation 111

5.5.2 The Number of Expressed Genes 111 5.5.3 Functions of some new genes expressed in the liver

and kidney of rifampicin treated and rifampicin plus stem

cells treated rats and their progenies 130 5.5.4 Analysis of Differential Gene Expression 139

5.6 Discussion 173 5.7 Conclusion 180

6 SUMMARY, GENERAL CONCLUSION AND

RECOMMENDATIONS FOR FUTURE RESEARCH 181

REFERENCES 182 APPENDICES 219 BIODATA OF STUDENT 424

LIST OF PUBLICATIONS 425

© COPYRIG

HT UPM

xv

LIST OF TABLES

Table Page

2.1 Variation in Expression of Immunophenotyping Surface Markers

Antibodies on Bone MarrowMesenchymal Stem Cell in Rats and

Mice as Compared with Human 22

2.2 Clinical Test of MSCs Based Treatment as Categorized by Disease

Kinds 31

2.3 Clinical Tests of MSC as Categorised by Phase 31

3.1 Osteogenesis Induction Medium Preparation 53

3.2 Mesenchymal Stem Cell Expansion Medium Preparation 53

3.3 Adipogenesis Induction Medium Preparation 55

3.4 Adipogenesis Maintenance Medium Preparation 55

3.5 Mesenchymal Stem Cell Expansion Medium Preparation 56

3.6 Differentiation Schedule for Adipogenesis induction 57

3.7 Chondrogenic Differentiation (CD) Medium Preparation 57

4.1 Mean ± Blood biochemistry Parameters of Rat and their progeny after

4-month treatment with rifampicin and rifampicin plus bone marrow

derived mesenchymal stem cells 70

4.2 Mean ± Quantitative histopathological analysis of kidney changes due

to rifampicin treatment and rifampicin plus bone marrow derived

mesenchymal stem cells 82

4.3 Mean ± Quantitative histopathological analysis of liver changes due to

rifampicin treatment and rifampicin with bone marrow derived

mesenchymal stem cells 87

5.1 First Strand Reaction Buffer and Random Primer Mix Preparation 105

5.2 Synthesis of First Strand cDNA 106

5.3 Synthesis of Second Strand cDNA 106

5.4 cDNA Library End Prep 107

© COPYRIG

HT UPM

xvi

5.5 Adaptor Ligation 108

5.6 Enrichment of PCR Library 109

5.7 PCR cycling conditions for Library Enrichment 109

5.8 Number of new genes from Liver of rifampicin treated adult rats 112

5.9 Number of new genes from Liver of rifampicin plus stem cells treated

adult rats 112

5.10 Number of new genes from Liver progeny of rifampicin treated rats 113

5.11 Number of new genes from Liver progeny of rifampicin plus stem

cells treated rats 115

5.12 Number of genes expressed in control progeny group but not

expressed in progeny of rifampicin treated Liver group 116

5.13 Number of genes expressed in control progeny group but not

expressed in progeny of rifampicin plus stem cells treated Liver group 118

5.14 Number of new genes from kidney of rifampicin treated adult rats 119

5.15 Number of new genes from kidney of rifampicin plus stem cells

treated adult rats 119

5.16 Number of new genes from kidney progeny of rifampicin treated rats 120

5.17 Number of new genes from kidney progeny of rifampicin plus stem

cells treated rats 121

5.18 Number of gene expressed in control group but not expressed in

rifampicin treated kidney rats 122

5.19 Number of gene expressed in control group but not expressed in

rifampicin plus stem cells treated kidney rats 122

5.20 Number of genes expressed in control progeny group but not

expressed in progeny of rifampicin treated kidney group 122

5.21 Number of genes expressed in control progeny group but not

expressed in progeny of rifampicin plus stem cells treated kidney

group 126

5.22 Statistical power to detect differential expression varies with effect

size, sequencing dept and number of replicates 139

© COPYRIG

HT UPM

xvii

5.23 Shows the expression levels of some fifty up-regulated genes between

male adult rifampicin treated liver and also male adult rifampicin plus

stem cells treated liver samples 140

5.24 Shows the expression levels of some fifty down-regulated genes

between male adult rifampicin treated liver and also male adult

rifampicin plus stem cells treated liver samples 143

5.25 Shows the expression levels of some fifty up-regulated genes between

male progeny rifampicin treated and also male progeny rifampicin

plus stem cells treated liver samples 146

5.26 Shows the expression levels of some fifty down-regulated genes

between male progeny rifampicin treated and also male progeny

rifampicin plus stem cells treated liver samples 149

5.27 Gene expression changes between rifampicin and rifampicin plus stem

cells treated adult kidney 152

5.28 Gene expression changes between rifampicin and rifampicin plus stem

cell treated progeny kidney 154

5.29 Gene expression changes between rifampicin and rifampicin plus stem

cell treated adult liver 157

5.30 Gene expression changes between rifampicin and rifampicin plus stem

cells treated progeny liver 159

5.31 Shows the expression levels of some fifty up-regulated genes between

male adult rifampicin treated kidney and also male adult rifampicin

plus stem cells treated kidney samples 162

5.32 Shows the expression levels of some fifty down-regulated genes

between male adult rifampicin treated kidney samples and also male

adult rifampicin plus stem cells treated kidney samples 165

5.33 Shows the expression levels of some fifty up-regulated genes between

male progeny rifampicin treated kidney samples and also male

progeny rifampicin plus stem cells treated kidney samples 168

5.34 Shows the heatmap expression levels of some fifty down-regulated

genes between male progeny rifampicin treated kidney samples and

also male progeny rifampicin plus stem cells treated kidney samples 171

© COPYRIG

HT UPM

xviii

LIST OF FIGURES

Figure Page

1.1 Research outlook 6

1.2 Research outlook continued 7

1.3 Research outlook continued 8

1.4 Research outlook continued 9

3.1 Morphological features of bone marrow derived mesenchymal stem

cells 59

3.2 Immunophenotyping Characterization of bone marrow derived

mesenchymal stem cells 60

3.3 Osteoblast detection in bone marrow derived mesenchymal stem cell 61

3.4 Adipocyte detection in bone marrow derived mesenchymal stem cell 62

3.5 Chondrogenic detection in bone marrow derived mesenchymal stem 62

4.1 Mean±SD of the concentrations (U/L) of alanine aminotransferase 71

4.2 Mean±SD of the concentrations (U/L) of aspartate aminotransferase 72

4.3 Mean±SD of the concentrations (mg/dl) of total bilirubin 73

4.4 Mean±SD of the concentrations (g/dl) of total protein 74

4.5 Mean±SD of the concentrations (g/dl) of albumin 75

4.6 Mean±SD of the concentrations (mg/dl) of urea 76

4.7 Mean±SD of the concentrations (mg/dl) of creatinine 77

4.8 Mean±SD of the concentrations (mg/dl) of triglyceride 78

4.9 Mean±SD of the concentrations (mg/dl) of cholesterol 79

4.10 Mean±SD of the concentrations (mg/dl) of low density lipolipid

cholesterol 80

4.11 Mean±SD of the concentrations (mg/dl) of high density lipolipid

cholesterol 81

© COPYRIG

HT UPM

xix

4.12 Histological examination of kidney tissue from the parent rats 83

4.13 Histological examination of kidney tissue from progeny rats 85

4.14 Mean glomeruli size of rats and their progenies 86

4.15 Histological examination of liver tissue from the parent rats 88

4.16 Histological examination of liver tissue from progeny rats 90

4.17 Mean hepatocyte size of rats and their progenies 91

4.18 Mean hepatocyte count of rats and their progenies 92

4.19 Mean bi-nucleated hepatocyte count of rats and their progenies 93

4.20 Mean Kupffer count of rats and their progenies 94

4.21 Mean pycnotic nuclei count of rats and their progenies 95

4.22 Mean vacuolated hepatocyte count of rats and their progenies 96

5.1 Gene expression differences of adult rat kidney trancriptomic profile 135

5.2 Gene expression differences of progeny rat kidney trancriptomic

profile 136

5.3 Gene expression differences of adult rat liver trancriptomic profile 137

5.4 Gene expression differences of progeny rat liver trancriptomic 138

5.5 The heatmap showing the expression levels of the first 50 upregulated

genes in adult liver 142

5.6 The heatmap showing the expression levels of the first 50

downregulated genes in adult liver 145

5.7 The heatmap showing the expression levels of the first 50 upregulated

genes in progeny 148

5.8 The heatmap showing the expression levels of the first 50

downregulated genes in progeny liver 151

5.9 The heatmap showing the expression levels of the first 50 upregulated

genes in adult kidney 164

5.10 The heatmap showing the expression levels of the first 50

downregulated genes in adult kidney 167

© COPYRIG

HT UPM

xx

5.11 The heatmap showing the expression levels of the first 50 upregulated

genes in progeny kidney 170

5.12 The heatmap showing the expression levels of the first 50

downregulated genes in progeny kidney 173

© COPYRIG

HT UPM

xxi

LIST OF ABBREVIATIONS

% Percentage

≤ Less than or equal

≥ Greater than or equal

µg Microgram

µL Micro-litre

µm Micrometer

µM Micromolar

A/G Albumin/Globulin

ACE Angiotensin-converting enzyme

AFB1 Aflatoxin B1

AGVHD Acute graft-versus-host disease

AIH Autoimmune hepatitis

AKI Acute kidney injury

ALD Adrenoleukodystrophy

ALF Acute liver failure

ALT Alanine amino transferase

ANOVA Analysis of variance

AP Acute pancreatitis

ARBs Angiotensin receptor blockers

ARF Acute renal failure

AST Aspatate amino transferase

BCL2 B-cell lymphoma 2

BDEC Bile duct epithelial cells

BDNF Brain-derived neurotrophic factor

BFGF Basic fibroblast growth factor

BM Bone marrow

BM-MNCs Bone marrow mononuclear cells

BM-MSC Bone marrow derived mesenchymal stem cells

Bw Between

© COPYRIG

HT UPM

xxii

C57BL/6 Black 6 mouse from Charles River Laboratories

CCl4 Carbon tetrachloride

CCR2 C-C chemokine receptor type 2

CCR3 C-C chemokine receptor type 3

CCR4 C-C chemokine receptor type 4

CCR5 C-C chemokine receptor type 5

CD Cluster of differentiation

CD Crohn’s disease

CGVHD Chronic graft-versus-host disease

CKD Chronic kidney disease

CLD Chronic liver disease

Cm2 Centimeter square

CPS Calcium phosphosilicate

CXCR4 Chemokine receptor type 4

DCs Dendritic cells

DEG Differentially expressed genes

DILI Drug-induced liver injury

DMEM Dulbecco's Modified Eagle's Medium

DMSO Dimethyl sulfoxide

DPBS Dulbecco's phosphate-buffered saline

ECM Extracellular molecule

EDTA Ethylenediaminetetraacetic acid

EGF Epidermal growth factor

EPO Erythropoietin

ESC Embryonic stem cell

ESC Embryonic stem cells

F1 generation Filial generation

FBS Fetal bovine serum

FDA Food and Drug Administration

FDR False discovery rate

FGF-4 Fibroblast growth factor 4

© COPYRIG

HT UPM

xxiii

FHF Fulminant hepatic failure

FHF Fulminant hepatic failure

Fig Figure

Foxp3 Forkhead box protein P3

FPKM Fragments Per Kilobase of transcript per Million

mapped reads

G Gram

G/dl Gram/deciliter

Ga1N Galactosamine induced

GFR Glomerular filteration rate

GO Gene ontology

HBV Hepatitis B virus

HCC Hepatocellular carcinoma

HCl Hydrochloric acid

HCV Hepatitis C virus

HDL High density lipolipid

HDL High density lipolipid

HGF Hepatocyte growth

HIV Human immunodeficiency virus

HLA Human leukocyte antigen

HLA-DR Human Leukocyte Antigen – antigen D Related

HLA-G5 Human leukocyte antigen G isoform

Hrs hour

HSC Hepatic stallate cells

I/R Ischemia-reperfusion

IACUC Institutional Animal Care and Used Committee

IGF-1 Insulin-like growth factor 1

IHC Immunohistochemistry

IL-10 Interleukin-10

IL-12 Interleukin-12

INR International normalize ratio

© COPYRIG

HT UPM

xxiv

IPF Idiopathic pulmonary fibrosis

IPSC Induced pluripotent stem cells

IU/L International unit per litre

KC Kupffer cells

KEGG Kyoto encyclopedia of genes and genomes

Kg Kilogram

LDL Low density lipolipid

LDL Low density lipolipid

LFT’s Liver function tests

LL 37 Cathelicidin antimicrobial peptide

LTBI Latent TB Infection

LVEF Left ventricular ejection fraction

M Molar

MCP-1 Monocyte chemo-attractant protein-1

MDR/RR-TB Multidrug-resistant TB/ Rifampicin-resistance TB

MDR-TR Multidrug-resistant TB

Mg Milligram

Mg/dl Milligram/deciliter

MI Myocardial infarction

Min Minute

ML Milliter

mM Millimolar

MMP3 Matrix metalloproteinase-3

MMP9 Matrix metalloproteinase-9

mRNA Messenger ribonucleic acid

MSC Mesenchymal stem cell

Mtb Mycobacterium tuberculosis

MR Male rifampicin

MC Male control

MRSC Male rifampicin plus stem cell

MDC Madin-Darby canine kidney epithelial cell

© COPYRIG

HT UPM

xxv

N Normality

NASH Nonalcoholic steatohepatitis

NCBI National Center for Biotechnology Information

NGS Next generation sequencing

NHPTK Human proximal tubular eprthelial cell

NK cell Natural killer cell

Nm Nanomolar

NSAIDs Non-steroidal anti-inflammatory drugs

oC Degree centigrade

OLT Orthotopic liver transplantation

PBC Primary biliary cirrhosis

PBS Phosphate buffered saline

PBSCT Peripheral blood stem cell transplantation

PCI Percutaneous coronary intervention

PCR Polymerase chain reaction

PDGF Platelet-derived growth factor

Pen-Strep Penicillin-streptomycin

PGE2 Prostaglandin-E2

PH Partial hepatectomy

PIGF Placenta growth factor

QC Quality checking

RNA Ribonucleic acid

RPM Revolution per minute

RR-TB Rifampicin-resistance TB

SD Standard deviation

SEC Sinusoidal endothelial cells

SGOT Serum glutamic oxaloacetic transaminase

SGPT Serum glutamate transaminase

STAT3 Signal transducer and activator of transcription 3

STAT3 Signal transducer and activator of transcription 3

STZ Streptozocin

© COPYRIG

HT UPM

xxvi

TB Tuberculosis

TCA Tricarboxylic acid

TE Tris EDTA

TGFα Transforming growth factorα

TGFβ1 Transforming growth factorβ-1

TNF tumor necrosis factor

U/L Units per litre

ULN Upper limit of normal

UPM Universiti Putra Malaysia

VEGF Endothelial growth factor

VEGF vascular endothelial growth factor

WHO World health organization

XDR-TB Extensive drug-resistance

ZDF Zucker diabetic fatty

© COPYRIG

HT UPM

1

CHAPTER 1

1 INTRODUCTION

1.1 Background

Infections caused by bacteria have become a main health predicament in recent

years, because some of the bacterial infections are very difficult to eradicate,

consequently leading to an increase in morbidity and mortality in both economically

advanced and less advanced countries (Samuel et al. 2011). Particularly, the disease

tuberculosis kills nearly 2 million people globally annually (Gursimrat et al. 2011,

Philippe et al. 2006). Although antibiotics clears off infection and improve the

health, for some infections, the courses of medication preceded up to several months

and do not completely eradicate the infection, which often reoccurs years or decades

after the initial treatment (Das et al. 2013).

Mycobacterium tuberculosis has a very puzzling method of pathogenesis. It seems to

defy logic by actually using macrophages, the front line defenses of the human

immune system, as a safe heaven for its reproduction. Also the bacteria can still

persist in secured niches like bone marrow mesenchymal stem cells, and then

reactivate the disease (Menzies, Al Jahdali and Otaibi, 2011), leading to therapeutic

failure (WHO, 2014). Furthermore, the bacteria has a spectacular ability of long term

persistence despite vigorous host immunity and prolonged therapy (Lonnroth and

Raviglione, 2008; Lilleback et al; 2003). The spread and bordened of tuberculosis

(TB) is one of the most present emergencies currently facing the human population,

with the untreated cases and multidrug resistant strains often leading to mortality.

Although frequencies of infection are decreasing with the introduction of antibiotics,

TB infection is once more increasing and currently 26% (2.7 million) of all

avoidable deaths in the world are due to TB (brown.edu, 2000). The global burden of

TB drug resistance as at 2015, were estimated at 480,000 new cases of multidrug-

resistant TB (MDR-TB) and additional 100,000 people with rifampicin-resistance

TB(RR-TB) who were also newly eligible for MDR-TB treatment. Drug resistance

surveillance data show that 3.9% of new and 21% of previously treated TB cases

was estimated to have had rifampicin-or multidrug-resistant tuberculosis (MDR/RR-

TB) in 2015. As in 2014, MDR-TB accounts for 3.3% of new TB cases. MDR/RR-

TB caused 250,000 deaths in 2015. Most cases and deaths occurred in Asia. About

9.5% of MDR-TB cases have additional drug-resistance, extensive drug-resistance

TB (XDR-TB). To date, 117 countries worldwide have at least one XDR-TB cases

(WHO, 2016).

TB is spread through breathing of airborne Mycobacterium tuberculosis cells which

multiply in macrophages and within the large cystic tubercles they form-liquified

caseous tissue surrounded by infected macrophages (Bichun et al., 1996). These

reduce regular functioning of the lungs, leading to its rupture to spread the pathogen.

In order to achieve efficient cure of tuberculosis, the treatment normally last for a

© COPYRIG

HT UPM

2

period of 6-9months. These prolong rifampicin treatment due to Mycobacterium

tuberculosis infection can leads to toxic effect and some vital organ damage.

Rifampicin being a bactericidal antibiotic drug in rifamycin group, semisynthetic

that originated from Streptomyces species, is the most efficient, effective and

considered a first line drug in the management and treatment of tuberculosis

worldwide (Eminzade et al., 2008), but its normally comes with several

toxicological side effects. Possible complications of tuberculosis and prolonged

rifampicin treatment include liver and kidney damage; these conditions can lead to

reduced efficiency of the affected organs and consequently lead to other diseases.

Rifampicin do generates a lot of morphological changes in the liver by its metabolic

activity, since the liver serves as the detoxification point of the drug (Santhosh et al.,

2007). There was a remarkable increase in lipid peroxidation in a four weeks

medication using rifampicin (20mg/kg) intraperitoneally as reported by Upadhyay et

al., 2007. An extraordinary elevation in triglycerides and cholesterol levels in rats

treated with rifampicin at a dose of 250mg/kg/day, for a period of one month was

noticed (Tasduq et al., 2007). In 2006, Santhosh et al, reported a remarkable

elevation in triglycerides, cholesterol and free fatty acids in serum sample of rats that

received (200mg rifampicin + 200mg isoniazid) for a period of one month. Pal et al.

(2008) reported a small decrease in the level high density lipolipid (HDL)

cholesterol with concurrent increase in low density lipolipid (LDL) cholesterol level,

while a remarkable elevation in serum sample of aspartate aminotransferase (AST)

and alanine aminotransferase (ALT) levels due to anti-TB medication was reported

by Rana et al., 2010.

Increase in levels of both AST and ALT were reported after treating patients

(Balamurugan et al., 2009) and mice with rifampicin (Upadhyay et al., 2007). A

decrease in albumin level was reported in tuberculosis patients and healthy

volunteers after studying the pharmacokinetics of the drug, because rifampicin

usually binds to albumin only (Rafiq et al., 2010). A reduction in total protein and

albumin levels in rat serum was noted after treating them with anti-TB drugs

(Santhosh et al., 2007 and Eminzade et al., 2008). Treatment of rats with rifampicin

at a dose of 10mg/kg/day for a period of 3-weeks revealed a remarkable reduction of

total bilirubin in the serum (Balamurugan et al., 2009). A remarkable rise in the

serum bilirubin, urea, and creatinine levels was reported in patients treated with

isoniazid (300mg/day), rifampicin (600mg/day), pyrazinamide (2g/day), and

ethanbutol (1g/day) for a period of eight months (Yanardag et al., 2005).

In a similar experiment, a remarkable rise in serum bilirubin and a reduction in

serum urea levels were recorded in rats treated with isoniazid and rifampicin 200mg

each/kg bw/day for a period of one month. Also an extraordinary elevation in serum

bilirubin and urea was reported, but with no meaningful changes in creatinine level

of rats that received 250mg/kg of rifampicin for a period of 4-weeks. The liver and

kidney toxicity of rifampicin treatment have also been reported by some researchers

like Awodele et al., 2010 and Rehka et al., 2005. Shabana et al. (2012) in a similar

experiment reported a remarkable elevation in total cholesterol, triglycerides and

© COPYRIG

HT UPM

3

LDL-cholesterol levels and with a reduction in HDL-cholesterol level. Also an

elevation in AST, ALT, bilirubin and urea was reported by Shabana et al., 2012,

with a drastic reduction in total protein, albumin and alpha 1-globulin and no

remarkable changes in globulin fractions (alpha 2-, beta- and gamma-globulin) and

albumin/globulin ratio (A/G) as well as creatinine level (Shabana et al., 2012).

Histopathological changes in liver like necrosis of hepatocytes, cytoplasmic

vacuolation, and distended sinusoids, lymphatic aggregations, while kidney damages

like glomeruli increased, mesangial matrix expansion and renal tubules regeneration

were also reported (Shabana et al., 2012) in albino rats that were treated with

rifampicin for a period of 4-weeks at a dose of 200mg/kg body weight/day via oral

gastric tubes (Santhosh et al., 2007). A lot of histopathological changes like; isolated

steatosis (through varying degrees of portal inflammation with neutrophils or

mononuclear infiltrate) to confluent necrosis in some patients that showed abnormal

liver function test within six weeks of rifampicin therapy were also reported

(Scheuer et al., 1974).

Also diffuse microvesicular fatty infilteration accompanied with mild portal triatidis

was reported in rats that received isoniazid treatment at a dose of 50mg/kg/day and

rifampicin 100mg/kg/day for a period of one week (Kalra et al., 2007). Furthermore,

combination of anti-TB drug: (100mg/kg/day + 50mg/kg/day + 100mg/kg/day of

pyrazinamide) once daily for a period of 3 months revealed membrane disintegration

and loss of the polyhedral structure in the liver, and also other abnormalities such as

necrosis, macro vesicular steatosis and inflammation were also reported. Prolong

rifampicin therapy can also cause hemolysis and subsequently acute kidney failure,

can also leads to interstitial nephritis (which is due to its direct toxic effect) and is

seen as part of pan-nephropathy (Lee, 1978). Renal lesion were seen, that are due to

the formation of immune complexes that were detected on capillary glomerular

basement membranes using the immunofluorescent and electron microscopy.

Deterioration in kidney activity seems to be acute in the case of reintroduction of

rifampicin (Covic et al., 1998).

The advancement in science and technology has led to the invention of stem cell, a

new concept for the regeneration of damaged tissues. Several reports on the success

of stem cell therapy and its practical application in regeneration of damaged tissues

prompted us to perform a preclinical study to determine its potential in managing the

tissue damages caused by prolonged rifampicin treatment. Stem cell therapy as a

regenerative form of medicine can be used in conjunction with the rifampicin to

manage the toxicological side effects and also to avert the architectural organ

damage. Bone marrow derived mesenchymal stem cells can differentiate into so

many cell types including the liver and kidney and there are ample evidences

indicating tissue repair or regeneration by differentiation of stem cells into the

damaged cell types. Thus, this present study will address the genes dysregulatory

conflict due rifampicin treatment and it is our hope that the damaged organs (kidney

and liver) due to prolong rifampicin treatment will be cured after receiving the stem

cell therapy.

© COPYRIG

HT UPM

4

1.2 Statement of the Problem

i. Rifampicin treatment due to Mycobacterium tuberculosis infection can caused

damage to some vital, such as liver and kidney. This can lead to acute liver

and kidney failure. Stem cell therapy as a regenerative form of medicine

using bone marrow derived mesenchymal stem cells can be used in

conjunction with the rifampicin to avert the organ damage and thereby

rescuing the affected organs.

1.3 Justification for the Study

i. Prolong rifampicin therapy due to tuberculosis and the toxicity of the drug;

coupled with the proportionate risk of kidney/liver malfunction has stress the

need for a new interventional approach. Intravenous administration of bone

marrow derived mesenchymal stem cells along with rifampicin can yield a

promising result. Because hepatocytes and renal cells growth factors that are

released by mesenchymal stem cells have multiple function of antiapoptotic

stimulation and antioxidant actions, as such can neutralized the toxicological

impacts of the drug. Also the ability of the transplanted MSCs to differentiate

into liver and kidney cells can help in the organ regeneration as well.

1.4 Hypothesis

i. Rifampicin treatment due to tuberculosis infection can cause damage to liver and

kidney and this can lead to alteration in the genetic pathways and also in the

chromosomes.

ii. These disorders may be carried to the next generations. So in our hypothesis

stem cell therapy can avert the organ damage caused by the rifampicin treatment

and may correct the dysregulated genes.

1.5 General Objective

To determine the therapeutic potentials of bone marrow derived mesenchymal

stem cells in averting organ damage due to rifampicin treatment in wistar rats

and their progenies.

Specific Objectives

i. To isolate, expand and characterized bone derived mesenchymal stem cells

from wistar rat.

ii. To determine the effectiveness of transplanted bone marrow derived

mesenchymal stem cells in averting organ damage due to rifampicin

treatment.

© COPYRIG

HT UPM

5

iii. To detect any differentially expressed genes (dysregulated genes) due to

prolong rifampicin treatment on the wistar rats and their progenies using next

generation sequence technique.

iv. To determine the effectiveness of transplanted bone marrow derived

mesenchymal stem cells in correcting the differentially expressed genes

(dysregulated genes) on Wistar rats and their progenies using next generation

sequence technique.

1.6 Research outlook

The research work involving animal experimentation is summarized into four parts

(Fig. 1.1, 1.2, 1.3, 1.4). The first part involves the oral administration of therapeutic

doses of rifampicin (9 mg/kg/day) to both male and female wistar rats while the

control received normal saline for the period of three consecutive months. The

second part involves breeding of the rats that were previously treated with rifampicin

(while the treatment continue during the breeding), after their delivery, blood, kidney

and liver samples were collected from the parents and their progenies for

biochemistry analysis, histology and RNA (Ribonucleic acid) extraction from liver

and kidney for next generation sequencing. The third part involves oral

administration of therapeutic doses of rifampicin 9mg/kg/day for 3-months and

intravenous administration of bone marrow derived mesenchymal stem cells, 100μl

2.5 x 105 cells twice/month for 3-months to both male and female wistar rat. The

fourth part involves breeding of the rats that were previously treated with rifampicin

and MSCs (while the treatment continue during the breeding), after their delivery,

blood, kidney and liver samples were collected from the parents and their progenies

for biochemistry analysis, histology and RNA extraction form liver and kidney for

next generation sequencing.

© COPYRIG

HT UPM

6

Figure 1.1 : Research outlook.

© COPYRIG

HT UPM

7

Figure 1.2 : Research outlook continued.

© COPYRIG

HT UPM

8

Figure 1.3 : Research outlook continued.

© COPYRIG

HT UPM

9

Figure 1.4 : Research outlook continued.

© COPYRIG

HT UPM

182

7 REFERENCES

Aggarwal, S. and Pittenger, M.F. (2005). Human mesenchymal stem cells modulate

allogeneic immune cell responses. Blood. 105: 1815-1822.

Agnihotri, M.S., Bansal, S. and Kumar, A. (1990). Acute renal failure due to

rifampicin. Indian J Chest Dis Allied Sci; 32(2): 125-128.

Alfarano, C., Roubeix, C., Chaaya R. et al., (2012). Intraparenchymal injection of

bone marrow mesenchymal stem cells reduces kidney fibrosis after ischemia-

reperfusion in cyclosporineimmunosuppressed rats. Cell Transplant, 21(9):

2009-2019.

Alison, M.R., Islam, S. and Lim, S. (2009). Stem cells in liver regeneration, fibrosis

and cancer: the good, the bad and the ugly. J Pathol., 217:282-298.

Am Esch, J.S., Knoefel, W.T., Klein, M. et al. (2005). Portal application of

autologous CD133+ bone marrow cells to the liver: a novel concept to

support hepatic regeneration. Stem Cells. 23: 463-470.

Amacher, D. (1998). Serum transaminase elevations as indicators of hepatic injury

following the administration of drugs. Regul Toxicol Pharmacol., 27: 119-

130.

Amado, L. C., Saliaris, A. P., Schuleri, K. H., et al., (2005). Cardiac repair with

intramyocardial injection of allogeneic mesenchymal stem cells after

myocardial infarction. Proceedings of the National Academy of Sciences of

the United States of America, 102(32): 11474-11479.

American Society of Health-Systems Pharmacists (2005). American Hospital

Formulary Service (AHFS) drug information [CD-ROM]. Gerald K.

McEvoy, ed. Marlborough, MA: Skyscape.

Amr, A. and Alaa, A.H., (2005): Oxidative stress mediates drug-induced

hepatotoxicity in rats: a possible role of DNA fragmentation. Toxicology,

208: 367-375.

Ando, Y., Inaba, M., Sakaguchi, Y. et al., (2008). Subcutaneous adipose tissue-

derived stem cells facilitate colonic mucosal recovery from 2,4,6-

trinitrobenzene sulfonic acid (TNBS)-induced colitis in rats. Inflammatory

Bowel Diseases, 14(6): 826-838.

Anglani, F., Forino, M., Del Prete, D., Tosetto, E., Torregrossa, R. and Angelo, A.

D. (2004). In search of adult renal stem cells. Journal of Cellular and

Molecular Medicine, 8(4): 474-487.

Appel, G.B. (2002). Tubulointerstitial diseases: drug-induced chronic interstitial

nephritis. ACP Medicine Online. New York, NY: WebMD; http://

www.medscape.com/viewarticle/534689. Accessed November 8, 2007.

© COPYRIG

HT UPM

183

Aronson, J.K. (2003): Drugs and renal insufficient. Medicine, 1:103-109.

Asari, S., Itakura, S., Ferreri, K., Liu, C.P., Kuroda, Y., Kandeel, F. and Mullen, Y.

(2009). Mesenchymal stem cells suppress B-cell terminal differentiation. Exp

Hematol., 37: 604-615.

Augello, A., Tasso, R., Negrini, S.M., Amateis, A., Indiveri, F., Cancedda, R. and

Pennesi, G. (2005). Bone marrow mesenchymal progenitor cells inhibit

lymphocyte proliferation by activation of the programmed death 1 pathway.

Eur J Immunol., 35: 1482-1490.

Awodele, O., Akintonwa, A., Osunkalu, V.O. and Coker, H.A.B. (2010).

Modulatory activity of antioxidants against the toxicity of rifampicin in vivo.

Rev. Inst. Med. Trop. São Paulo., 52(1): 43-46.

Azimifar, S. B., Nagaraj, N., Cox, J. and Mann, M. (2014). Cell-type-resolved

quantitative proteomics of murine liver, Cell Metabolism, 20(6):1076–1087.

Bachs, L., Pares, A., Elena, M., Piera, C. and Rodes, J. (1992): Effects of long-term

rifampicin administration in primary biliary cirrhosis. Gastroenterology;

102:2077-2080.

Bagul, A., Frost, J.H. and Drage, M. (2013): Stem cells and their role in renal

ischaemia reperfusion injury. Am J Nephrol.; 37: 16-29.

Baksh, D., Song, L. and Tuan, R.S. (2004): Adult mesenchymal stem cells:

characterization, differentiation, and application in cell and gene therapy. J

Cell Mol Med. 8:301-316.

Balamurugan, K., Vanithakumari, G. and Indra, N. (2009). Effect of rifampicin on

certain biochemical parameter in the liver of albino rats. Internet J. Toxicol.,

6(1).

Barnes, N., Bellamy, D., Ireland, L., et al. (1984). Pulmonary tuberculosis

complicated by haemophagocyte syndrome and rifampicininduced

tubulointerstitial nephritis. Br J Dis Chest., 78: 395-397.

Barry, F.P. and Murphy, J.M. (2004). Mesenchymal stem cells: clinical applications

and biological characterization. Int J Biochem Cell Biol., 36: 568-584.

Barzilay, R., Sadan, O., Melamed, E. and Offen, D. (2009). Comparative

characterization of bone marrow-derived mesenchymal stromal cells from

four different rat strains. Cytotherapy, 11(4):435–442.

Basaraba, R.J. (2008). Experimental tuberculosis: the role of comparative pathology

in the discovery of improved tuberculosis treatment strategies. Tuberculosis

(Edinb)., 88: Suppl 1S35-47.

Bataller, R. and Brenner, D. A. (2005). Liver fibrosis. Journal of Clinical

Investigation, 115(2): 209-218.

© COPYRIG

HT UPM

184

Bellomo, R. (2006). The epidemiology of acute renal failure: 1975 versus 2005.

Curr Opin Crit Care., 12(6): 557-560.

Benichou, C. (1990). Criteria for drug-induced liver disorder: report of an

international consensus meeting. J Hepatol., 11:272-276.

Benson, E.A., Eadon, M.T., Desta, Z., Liu, Y., Lin, H., Burgess, K.S., Segar, M.W.,

Gaedigk, A. and Skaar, T.C. (2016). Rifampin Regulation of Drug

Transporters Gene Expression and the Association of MicroRNAs in Human

Hepatocytes. Frontier in Pharmacology, 7(111): 1-13.

Bexell, D., Gunnarsson, S., Tormin, A. et al. (2009). Bone marrow multipotent

mesenchymal stroma cells act as pericytelike migratory vehicles in

experimental gliomas. Molecular Therapy, 17(1):183–190.

Bhattacharya., H., Lun, L. and Gomez, G.D. (2005). Biochemical effects to toxicity

of CCL4 on rosy barbs (Puntius conchonius). Our Nature, 3: 20-25.

Bi, B., Schmitt, R. and Israilova, M. et al., (2007). Stromal cells protect against acute

tubular injury via an endocrine effect. J Am Soc Nephrol.,18: 2486-2496.

Bichun, L.A., Pedrosa, M.M., Trippel, S.J. (1996). Molecular Biology of

mycobacterium tuberculosis.

<http://www.sp.uconn.edu/~terry/Spring96/WebTB2/Groups/Group14/Final.

html> Accessed 2000 Apr. 20.

Bihorac, A., Ozener, C., Akoglue, E. and Kullu, S. (1999). Tetracyclineinduced

acute interstitial nephritis as a cause of acute renal failure. Nephron, 81(1):

72-75.

Bishop-Bailey, D., Thomson, S., Askari, A., Faulkner, A. and Wheeler-Jones, C.

(2014). Lipid-metabolizing CYPs in the regulation and dysregulation of

metabolism. Annu. Rev. Nutr., 34: 261–279.

Bo¨cker, R., Estler, C.J., Mu¨ller, S., Pfandzelter, C. and Spachmu¨ller, B. (1982).

Comparative evaluation of the effects of tetracycline, rolitetracycline and

doxycycline on some blood parameters related to liver function. Arzneim-

Forsch. Drug Res., 32(3): 237-241.

Bouffi, C., Bony, C., Courties, G., Jorgensen, C. and Noel, D. (2010). IL-6-

dependent PGE2 secretion by mesenchymal stem cells inhibits local

inflammation in experimental arthritis. PLoS One. 5: e14247-10.1371.

Boulton-Jones., J.M., Sissons, J.G.P., Evans, D.J. and Peters, D.K. (1974). Renal

lesions of subacute infective endocarditis. Br. Med. J., 2: 11-14.

Brasaemle, D.L., Subramanian, V., Garcia, A., Marcinkiewicz, A., Rothenberg, A.

(2009). Perilipin a and the control of triacylglycerol metabolism. Mol. Cell.

Biochem., 326: 15–21.

© COPYRIG

HT UPM

185

Brazelton, T.R., Rossi, F.M., Keshet, G.I. and Blau, H.M. (2000). From marrow to

brain: Expression of neuronal phenotypes in adult mice. Science. 290: 1775-

1779.

Brenner, S., Whiting-Theobald, N., Kawai, T. et al., (2004). CXCR4-transgene

expression significantly improves marrow engraftment of cultured

hematopoietic stem cells. Stem Cells. 22: 1128-1133.

Brewer, C.T. and Chen, T. (2016). PXR variants: The impact on drug metabolism

and therapeutic responses. Acta Pharm. Sin. B, 6: 441–449.

Brown, S.J. and Desmond, P.V. (2002). Hepatotoxicity of antimicrobial agents.

Semin Liver Dis., 22: 157-167.

Brown.edu.TBgenome. (2000). <http://www.brown.edu/Research/TBHIV_Lab/tb/tbi

ntro.html> Accessed 2000 Apr 20.

Burk, O., Koch, I., Raucy, J., Hustert, E., Eichelbaum, M., Brockmoller, J., Zanger,

U.M. and Wojnowski, L. (2004). The induction of cytochrome P450 3A5

(CYP3A5) in the human liver and intestine is mediated by the xenobiotic

sensors pregnane X receptor (PXR) and constitutively activated receptor

(CAR). J Biol Chem., 279:38379-38385.

Byrne, J.A., Strautnieks, S.S., Mieli-Vergani, G., Higgins, C.F., Linton, K.J. and

Thompson, R.J. (2002): The human bile salt export pump: characterization of

substrate specificity and identification of inhibitors. Gastroenterology, 123:

1649-1658.

Cameron, J.S. (1988). Allergic interstitial nephritis: clinical features and

pathogenesis. Q J Med.,250: 97-102.

Capelle, P., Dhumeaux, D., Mora, M., Feldmann, G. and Berthelot, P. (1972). Effect

of rifampicin on liver function in man. Gut, 13: 366-371.

Capuano, S.V. 3rd., Croix, D.A., Pawar, S., Zinovik, A., Myers, A. et al. (2003).

Experimental Mycobacterium tuberculosis infection of cynomolgus

macaques closely resembles the various manifestations of human M.

tuberculosis infection. Infect Immun 71: 583-5844.

Caroll, N. (1987). Tuberculous hypercalcaemia with renal failure. Br J Dis Chest.,

81(3): 296-299.

Carro Mendez, B., Rozas Lozano, P., Esteban Morcillo, J. and Otero Gonzales, A.

(1980). Acute kidney failure (AKF) and hemolysis second- ary to accidental

discontinuous treatment with rifampicin. Anales de Medicina Interne., 8(10):

521.

Carlton, H.M. (1980). Carlton’s histological techniques, 5th edition, Churchill,

Living stone, Edinburgh.

© COPYRIG

HT UPM

186

Carson, J.L., Strom, B.L., Duff, A., Gupta, A. and Das, K. (1993). Safety of

nonsteroidal anti-inflammatory drugs with respect to acute liver disease. Arch

Intern Med., 153: 1331-1336.

Cascio, A., Scarlata, F., Giordano, S., Antinori, S., Colomba, C. and Titone, L.

(2003). Treatment of human brucellosis with rifampin plus minocycline. J

Chemother; 15: 248-252.

Castelo-Branco, M. T., Soares, I. D., Lopes, D. V. et al., (2012). Intraperitoneal but

not intravenous cryopreserved mesenchymal stromal cells home to the

inflamed colon and ameliorate experimental colitis. PLoS One, 7(3) Article

ID e33360.

Chan, W.C., O’Mahoney, G., Sister, M. Yu Ryh. and Yu, Dyo. (1975). Renal failure

during intermittent rifampicin therapy. Tubercle., 56: 191–194.

Chen, L., Zhang, W., Yue, H., Han, Q., Chen, B., Shi, M., Li, J., Li, B., You, S. and

Shi, Y. (2007). Effects of human mesenchymal stem cells on the

differentiation of dendritic cells from CD34+ cells. Stem Cells Dev., 16: 719-

731.

Chaudhary, J.C. and Rath, P.C. (2017). A simple method for isolation, propagation,

characterization, and differentiation of adult mouse bone marrow-derived

multipotent mesenchymal stem cells. Journal of cell science and therapy.

8:261.

Chen, S.L., Fang, W.W., Ye, F., Liu, Y.H., Qian, J., Shan, S.J., Zhang, J.J.,

Chunhua, R.Z., Liao, L.M. and Lin, S. (2004). Effect on left ventricular

function of intracoronary transplantation of autologous bone marrow

mesenchymal stem cell in patients with acute myocardial infarction. Am J

Cardiol., 94: 92-95.

Cheng, J.T. and Kahn, T. (1984). Potassium wasting and other renal tubular defects

with rifampin nephrotoxicity. Am J Nephrol., 4: 379–382.

Cheng D., Nelson T.C., Chen J., Walker S.G., Wardwell-Swanson J., Meegalla

R., Taub R., Billheimer J.T., Ramaker M. and Feder J.N. (2003).

Identification of acyl coenzyme A:monoacylglycerol acyltransferase 3, an

intestinal specific enzyme implicated in dietary fat absorption. J. Biol. Chem.

278:13611-13614.

Cheng, X., and Klaassen, C. D. (2006). Regulation of mRNA expression of

xenobiotic transporters by the pregnane x receptor in mouse liver, kidney,

and intestine. Drug Metab. Dispos. 34, 1863–1867.

Chitturi, S. and Farrell, G. (2002). Drug-induced liver disease. In: Schiff ER, Sorrell

MF, Maddrey WC, editors. Schiff’s diseases of the liver, 9th ed.

Philadelphia: Lippincott, Williams & Wilkins, pp. 1059-1128.

© COPYRIG

HT UPM

187

Choi, S., Park, M., Kim, J., Hwang, S., Park, S. and Lee, Y. (2009). The role of

mesenchymal stem cells in the functional improvement of chronic renal

failure. Stem Cells Dev., 18: 52-529.

Christodoulus Xinaris, M., Morigi, V., Benedetti, B., Imberti, A.S., Fabricio, E.,

Squarcina, A., Beniggni, E. and Gagliardini, G. Remuzzi (2013):

Mesenchymal stem cell migration capacity and promote tissue repair in an

injury specific fashion. Cell Transplant. 22: 423-436.

Clinicaltrials.gov.http://clinicaltrials.gov,

Coco, T.J. and Klasner, A.E. (2004). Drug-induced rhabdomyolysis. Curr Opin

Pediatr., 16(2): 206-210.

Cohn, J.R., Fye, D.L., Sills, J.M. and Francos, G.C. (1985). Rifampicin induced

renal failure. Tubercle, 66(4): 289-293.

Colter, D.C., Sekiya, I., Prockop, D.J. (2001). Identification of a subpopulation of

rapidly self-renewing and multipotential adult stem cells in colonies of

human marrow stromal cells. Proc Natl Acad Sci U S A. 98: 7841-7845.

Corcione, A., Benvenuto, F., Ferretti, E., Giunti, D., Cappiello, V., Cazzanti, F.,

Risso, M., Gualandi, F., Mancardi, G.L. and Pistoia, V. (2006). Human

mesenchymal stem cells modulate B-cell functions. Blood. 107: 367-372.

Couto, B.G., Goldenberg, R.C., Da Fonseca, L.M. et al. (2011). Bone marrow

mononuclear cell therapy for patients with cirrhosis: a Phase 1 study. Liver

Int., 31(3): 391-400.

Covic, V., Goldsmith, D.J., Segall, L., Stoicescu, C., Lungu, S., Volovat, C. and

Covic, M. (1998). Rifampicin-induced acute renal failure: a series of 60

patients. Nephrol. Dial. Transplant. 13(4): 924-929.

Culling.C.F.K. (1974). Handbook of Histopathological and Histochemical

Techniques, third Edition. Trowbridge an Esher publishers redwood burn

limited, pp. 712-730.

Dalgetty, D.M., Medine, C.N., Iredale, J.P. et al. (2009). Progress and future

challenges in stem cell-derived liver technologies. Am J Physiol Gastrointest

Liver Physiol., 297(2): G241-G248.

Dan, Y. and Yeoh, G. (2008). Liver stem cells: a scientific and clinical perspective. J

Gastroenterol Hepatol., 23: 687-698.

Dalen, K.T., Schoonjans, K., Ulven, S.M., Weedon-Fekjaer, M.S., Bentzen, T.G.,

Koutnikova, H., Auwerx, J. and Nebb, H.I. (2004). Adipose tissue expression

of the lipid droplet–associating proteins S3–12 and perilipin is controlled by

peroxisome proliferator—Activated receptor-γ. Diabetes, 53: 1243–1252.

© COPYRIG

HT UPM

188

Dan, Y.Y., Riehle, K.J., Lazaro, C., Teoh, N., Haque, J., Campbell, J.S. and Fausto,

N. (2006): Isolation of multipotent progenitor cells from human fetal liver

capable of differentiating into liver and mesenchymal lineages. Proc Natl

Acad Sci U S A, 103: 9912-9917.

Dannenberg, A.M. (1994): Rabbit model of tuberculosis. In: Bloom BR, editor.

Tuberculosis: pathogenosis, protection and control. Washington, SC: ASM

Press., pp. 135-147.

Das, B., Kashino, S.S., Pulu, I., Kalita, D., Swami, V., Yeger, H., Felsher, D.W and

Campos-Neto, A. (2013). CD271+ bone marrow mesenchymal stem cells

may provide a niche for dormant Mycobacterium tuberculosis. Sci Transl

Med 2013, 5:170ra113.

Davidson, M.B., Thakkar, S., Hix, J.K., Bhandarkar, N.D., Wong, A. and Schreiber,

M.J. (2004). Pathophysiology, clinical consequences, and treatment of tumor

lysis syndrome. Am J Med., 116(8): 546-554.

Delorme, B., Ringe, J., Gallay, N. et al. (2008). Specific plasma membrane protein

phenotype of culture-amplified and native human bone marrow mesenchymal

stem cells. Blood, 111(5): 2631–2635.

Delorme, B. and Charbord, P. (2007). Culture and characterization of human bone

marrow mesenchymal stem cells. Methods in Molecular Medicine, 140: 67–

81.

De Hemptinne, I., Vermeiren, C., Maloteaux, J.M. and Hermans, E. (2004).

Induction of glial glutamate transporters in adult mesenchymal stem cells. J

Neurochem 91:155-166.

Deng, J., Petersen, B.E., Steindler, D.A., Jorgensen, M.L. and Laywell, E.D. (2006).

Mesenchymal stem cells spontaneously express neural proteins in culture and

are neurogenic after transplantation. Stem Cells. 24: 1054-1064.

Di Nicola, M., Carlo-Stella, C., Magni, M., Milanesi, M., Longoni, P.D., Matteucci,

P., Grisanti S. and Gianni, A.M. (2002). Human bone marrow stromal cells

suppress t-lymphocyte proliferation induced by cellular or nonspecific

mitogenic stimuli. Blood., 99: 3838-3843.

Dielh, A.M. (2000): Cytokine regulation of liver injury and repair. Immunol Rev.,

174: 160-171.

Diczfalusy, U., Nylen, H., Elander, P. and Bertilsson, L. (2011). 4β-

Hydroxycholesterol, an endogenous marker of CYP3A4/5 activity in

humans. Br. J. Clin. Pharmacol., 71: 183–189.

Dominici, M., Hofmann, T.J. and Horwitz, E.M. (2001). Bone marrow mesenchymal

cells: biological properties and clinical applications. J Biol Regul Homeost

Agents., 15: 28-37.

© COPYRIG

HT UPM

189

Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F., Krause,

D., Deans, R., Keating, A., Prockop, D. and Horwitz, E. (2006). Minimal

criteria for defining multipotent mesenchymal stromal cells. The

International Society for Cellular Therapy position statement. Cytotherapy. 8:

315-317.

Dong, X. J., Zhang, H., R. Pan, L., Xiang, L. X. and Shao, J. Z. (2010).

Identification of cytokines involved in hepatic differentiation of mBM-MSCs

under liver-injury conditions. World Journal of Gastroenterology, 16(26):

3267-3278.

Donato, M., Jimenez, N., Serralta, A., Mir, J., Castell, J. and Gómez-Lechón, M.

(2007). Effects of steatosis on drug-metabolizing capability of primary

human hepatocytes. Toxicol. In Vitro, 21: 271–276.

Dufour, D.R., Lott, J.A., Nolte, F.S., Gretch, D.R., Koff, R.S. and Seeff, L.B. (2000).

Diagnosis and monitoring of hepatic injury: I. Performance characteristics of

laboratory tests. Clin Chem., 46: 2027-2049.

Dufour, D.R., Lott, J.A., Nolte, F.S., Gretch, D.R., Koff, R.S. and Seeff, L.B. (2000).

Diagnosis and monitoring of hepatic injury: II. Recommendations for use of

laboratory tests in screening, diagnosis, and monitoring. Clin Chem., 46:

2050-2068.

Duncan, A.W., Dorrell, C. and Grompe, M. (2009). Stem cells and liver

regeneration. Gastroenterology. 137: 466-481.

Ebrahimi, B., Eirin, A., Li, Z., Zhu, X.Y., Zhang, X., Lerman, A., Textor, S.C. and

Lerman, L.O. (2013). Mesenchymal stem cells improve medullary

inflammation and fibrosis after revascularization of swine atherosclerotic

renal artery stenosis. PLoS One, 8: e67474-10.1371.

Edalatmanesh, M. A., Bahrami, A. R., Hosseini, E., Hosseini, M. and Khatamsaz, S.

(2011). Bone marrow derived mesenchymal stem cell transplantation in

cerebellar degeneration: a behavioral study. Behavioural Brain Research,

225(1): 63-70.

Eirin, A., Zhu, X.Y., Krier, J.D., Tang, H., Jordan, K.L., Grande, J.P., Lerman, A.,

Textor, S.C. and Lerman, L.O. (2012). Adipose tissue-derived mesenchymal

stem cells improve revascularization outcomes to restore renal function in

swine atherosclerotic renal artery stenosis. Stem Cells, 30: 1030-1041.

Eminzade, S., Uras, F. and Izzettin, F.V. (2008): Silymarin Protects liver against

toxic effects of anti-tuberculosis drugs in experimental animals. Nutr. Metab.,

5: 18.

© COPYRIG

HT UPM

190

English, K., Ryan, J.M., Tobin, L., Murphy, M.J., Barry, F.P. and Mahon, B.P.

(2009). Cell contact, prostaglandin E(2) and transforming growth factor beta

1 play non-redundant roles in human mesenchymal stem cell induction of

CD4 + CD25(High) forkhead box P3+ regulatory T cells. Clin Exp Immunol.,

156: 149-160.

Enriquez-Cortina, C., Almonte-Becerril, M., Clavijo-Cornejo, D., Palestino-

Domínguez, M., Bello-Monroy, O., Nuño, N., López, A., Bucio, L., Souza,

V., Hernández-Pando, R. et al. (2013). Hepatocyte growth factor protects

against isoniazid/rifampicin-induced oxidative liver damage. Toxicol. Sci.,

135: 26–36.

Erdil, A., Kadayifci, A., Ates, Y., Bagci, S., Uygun, A. and Dagalp, K. (2001).

Rifampicin test in the diagnosis of Gilbert’s syndrome. Int J Clin Pract, 55:

81-83.

Ezquer, F.E., Ezquer, M.E., Parrau, D.B., Carpio, D., Yanez, A.J. and Conget, P.A.

(2008). Systemic administration of multipotent mesenchymal stromal cells

reverts hyperglycemia and prevents nephropathy in type 1 diabetic mice. Biol

Blood Marrow Transplant.14: 631-640.

Fang, X., Neyrinck, A.P., Matthay, M.A. and Lee, J.W. (2010). Allogeneic human

mesenchymal stem cells restore epithelial protein permeability in cultured

human alveolar type II cells by secretion of angiopoietin-1. J Biol Chem.,

285: 26211-26222.

Farrell, G.C. (1995). Drug-Induced Liver Disease. Churchill Living Stone, London,

pp. 413-430.

Farrell, G.C. (2002). Drugs and steatohepatitis. Semin Liver Dis., 22: 185-194.

Fausto, N. and Riehle, K.J. (2005). Mechanisms of liver regeneration and their

clinical implications. J Hepatobiliary Pancreat Surg, 12(3): 181-189.

Fausto, N., Campbell, J.S. and Riehle, K.J. (2006). Liver regeneration. Hepatology.

43(Suppl.1): S45-S53.

Fellous, T.G., Islam, S., Taudros, P.J. et al. (2009). Locating the stem cell niche and

tracing hepatocyte lineages in human liver. Hepatology. 49: 1655-1663.

Flax, M.H. and Waksman, B.H. (1962). Delayed cutaneous reactions in the rat. J

Immunol 89: 496-504.

Flynn, J.L. and Chan, J. (2003). Animal models of tuberculosis. In: Rom SMG,

editor. Tuberculosis. 2nd edn. Philadelphia: Lippincott, Williams & Wilkins,

pp. 237-250.

© COPYRIG

HT UPM

191

Flynn, J.L., Tsenova, L., Izzo, A. and Kaplan, G. (2008). Experimental Animal

Models of Tuberculosis. In: Kaufmann SH, Britton WJ, editors. Handbook of

Tuberculosis: Immunology and Cell Biology. Weinheim: WILEY-VCH

Verlag GmbH & Co. KGaA.

Foraker, J.E., Oh, J.Y., Ylostalo, J.H., Lee, R.H., Watanabe, J. and Prockop, D.J.

(2011). Cross-talk between human mesenchymal stem/progenitor cells

(MSCs) and rat hippocampal slices in LPS-stimulated cocultures: the MSCs

are activated to secrete prostaglandin E2. J. Neurochem., 119: 1052-1063.

Fored, C.M., Ejerblad, E., Lindblad, P., et al. (2001). Acetaminophen, aspirin, and

chronic renal failure. N Engl J Med., 345(25): 1801-1808.

Frank, M.H. and Sayegh, M.H. (2004). Immunomodulatory functions of

mesenchymal stem cells. Lancet., 363: 1411-1412.

Franquesa, M., Herrero, E., Torras, J., Ripoll, E., Flaquer, M., Goma, M., Lloberas,

N., Anegon, I., Cruzado, J.M., Grinyo, J.M. and Herrero-Fresneda, I. (2012).

Mesenchymal stem cell therapy prevents interstitial fibrosis and tubular

atrophy in a rat kidney allograft model. Stem Cells Dev., 21: 3125-3135.

Friedenstein, A.J., Petrakova, K.V., Kurolesova, A.I. and Frolova, G.P. (1968).

Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and

hematopoietic tissues. Transplantation. 6: 230-247.

Fujii, H., Ikura, Y., Arimoto, J., Sugioka, K., Iezzoni, J.C., Park, S.H., Naruko, T.,

Itabe, H., Kawada, N. and Caldwell, S.H. (2010). Expression of perilipin and

adipophilin in nonalcoholic fatty liver disease; relevance to oxidative injury

and hepatocyte ballooning. J. Atheroscler. Thromb., 16: 893–901.

Gabow, P.A., Lacher, J.W. and Neff, T.A. (1976). Tubulointerstitial and glomerular

nephritis associated with rifampicm. JAMA., 235: 2517-2518.

Gabriel, R. (1971): Rifampicin jaundice. BMJ., 3: 182.

Gabriele C. (2010). Genome-wide analysis of eukaryotic twin CX9C proteins. Mol.

BioSyst., 6:2459–2470.

Galal, A.M., Walker, L.A. and Khan, I.A. (2015). Induction of gst and related

events by dietary phytochemicals: Sources, chemistry, and possible

contribution to chemoprevention. Curr. Top. Med. Chem., 14: 2802–2821.

Gandhi, T.K., Burstin, H.R., Cook, E.F., et al. (2000). Drug complications in

outpatients. J Gen Intern Med.; 15(3): 149-154.

Geevasinga, N., Coleman, P.L., Webster, A.C. and Roger, S.D. (2006). Proton pump

inhibitors and acute interstitial nephritis. Clin Gastroenterol Hepatol., 4(5):

597-604.

© COPYRIG

HT UPM

192

Georgina A., Carmen E., Herena E, Luisa M. V., José C. A., Carmen P., MSc J.K.,

Giulio D., Ludwig K., Jaume S., Deborah P., Eva S., Manuel C., Jordi R.,

Carlos N., Carmen T., Joaquín C., Angela V., Ingrid G., Maria J. A., Cristina

A., Alex R., Xavier M. and Mar T. (2016). Neurofilament light chain level is

a weak risk factor for the development of MS. Neurology. 87:1076–1084.

Girling, D.J. (1973). Side-effects observed during intermittent rifampicin therapy.

Scand J Respir Dis., 84: 119-120.

Giacomini, K. M., Huang, S. M., Tweedie, D. J., Benet, L. Z., Brouwer, K. L., Chu,

X., et al. (2010). Membrane transporters in drug development. Nat. Rev.

Drug Discov. 9, 215–236.

Glyone, S.R and Page, D.S. (1923): The reaction to B. tuberculosis in the albino rat.

J Pathol Bacteriol; 26: 224–233.

Gomez, J.E. and McKinney, J.D. (2004). M. tuberculosis persistence, latency, and

drug tolerance. Tuberculosis, 84: 29-44.

Graham, D.J., Staffa, J.A., Shatin, D., et al. (2004). Incidence of hospitalized

rhabdomyolysis in patients treated with lipid-lowering drugs. JAMA.,

292(21): 2585-2590.

Gray, D.F. (1961). The relative natural resistance of rats and mice to experimental

pulmonary tuberculosis. J Hyg (Lond)., 59: 471-477.

Greenberg, A.S., Egan, J.J., Wek, S.A., Garty, N.B., Blanchette-Mackie, E. and

Londos, C. (1991). Perilipin, a major hormonally regulated adipocyte-

specific phosphoprotein associated with the periphery of lipid storage

droplets. J. Biol. Chem., 266: 11341–11346.

Gronhagen-Riska, C., Hellstrom, P.E. and Froseth, B. (1978). Predisposing factors in

hepatitis induced by isoniazid-rifampin treatment of tuberculosis. Am Rev

Respir Dis., 118: 461-466.

Grosset, J. and Leventis, S. (1983). Adverse effects of rifampin. Rev Infect Dis., 5:

S440-S450.

Grunfeld, J.P., Kleinknecht, D. and Droz, D. (1993). Acute interstitial nephritis. In:

Diseases of the Kidney. Little Brown, Boston, pp. 1331-1353.

Grusch, M., Drucker, C., Peter-Vorosmarty, B., Erlach, N., Lackner, A. et al. (2006).

Deregulation of the activin/follistatin system in hepatocarcinogenesis.

Journal of hepatology 45: 673–680.

Gupta, A., Sakhuja, V. and Chugh, K.S. (1992). Intravascular hemolysis and acute

renal failure following imtermittent rifampin therapy. Int J Lepr Other

Mycobact Dis., 60(2): 185-188.

© COPYRIG

HT UPM

193

Gupta, N., Su, X., Popov, B., Lee, J.W., Serikov, V. and Matthay, M.A. (2007).

Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells

improves survival and attenuates endotoxin-induced acute lung injury in

mice. J. Immunol., 179: 1855-1863.

Gursimrat, K. Sandhu. (2011): Tuberculosis: Current Situation, Challenges and

Overview of its Control Programs in India. J.global infect. Diseas., 3(2):

143-150.

Guzzini, F., Angiolini, F., Cazzaniga, L.,Gasparini, P., Milvio, E. and Mosconi, L.

(1994). Immune hemolytic anemia and acute kidney failure due to

rifampicin. Recenti Prog Med., 85(3): 182-185.

GTEx Consortium (2013). The Genotype-Tissue expression (GTEx) project. Nat

Genet. 45:580-585.

Harting, M. T., Jimenez, F., Pati, S., Baumgartner, J. and Cox, C. S. (2008).

Immunophenotype characterization of rat mesenchymal stromal cells.

Cytotherapy, 10(3):243–253.

Han, B., Lu, Y., Meng, B. et al. (2009). Cellular loss after allogenic hepatocyte

transplantation. Transplantation. 87: 1-5.

Hanioka, N., Nonaka, Y., Saito, K., Negishi, T., Okamoto, K. et al. (2012). Effect of

aflatoxin B1 on UDP-glucuronosyltransferase mRNA expression in HepG2

cells. Chemosphere 89: 526–529. 44.

Herbert, B. S., Grimes, B. R., Xu, W. M., Werner, M., Ward, C., Rossetti, S., et al.

(2013). A telomerase immortalized human proximal tubule cell line with a

truncation mutation (Q4004X) in polycystin-1. PLoS ONE 8:e55191. doi:

10.1371/journal.pone.0055191.

Herman, J.S. and Easterbrook, P.J. (2001). The metabolic toxicities of antiretroviral

therapy. Int J STD AIDS., 12: 555-562.

Herrera, M. B., Bussolati, B., Bruno, S. et al., (2007). Exogenous mesenchymal stem

cells localize to the kidney by means of CD44 following acute tubular injury.

Kidney International, 72(4): 430-441.

Hewitt, N.J., Gómez Lechón, M.J., Houston, J.B., Hallifax, D., Brown, H.S., Maurel,

P., Kenna, J.G., Gustavsson, L., Lohmann, C. and Skonberg, C. (2007).

Primary hepatocytes: Current understanding of the regulation of metabolic

enzymes and transporter proteins, and pharmaceutical practice for the use of

hepatocytes in metabolism, enzyme induction, transporter, clearance, and

hepatotoxicity studies. Drug Metab. Rev., 39: 159–234.

Hillgren, K. M., Keppler, D., Zur, A. A., Giacomini, K. M., Stieger, B., Cass, C. E.,

et al. (2013). Emerging transporters of clinical importance: an update from

the International Transporter Consortium. Clin. Pharmacol. Ther. 94, 52–63.

© COPYRIG

HT UPM

194

Hirsch, D.J., Bia, F.J., Kashgarian, M. and Bia, M.J. (1983). Rapidly progressive

glomerulonephritis during antituberculous therapy. Am J Nephrol., 3: 7-8.

Hong Kong Chest Service (1992). Tuberculosis Research Centre, Madras, British

Medical Research Council. A double-blind placebo-controlled clinical trial of

three anti-tuberculosis chemoprophylaxis regimens in patients with silicosis

in Hong Kong. Am Rev Respir Dis., 145: 36-41.

Horwitz, E. M., Gordon, P. L., Koo, W. K. K. et al., (2002). Isolated allogeneic bone

marrow-derived mesenchymal cells engraft and stimulate growth in children

with osteogenesis imperfecta: implications for cell therapy of bone.

Proceedings of the National Academy of Sciences of the United States of

America, 99(13): 8932-8937.

Horwitz, E.M. (2003). Stem cell plasticity: the growing potential of cellular therapy.

Arch Med Res., 34: 600-606.

Horwitz, E.M., Gordon, P.L., Koo, W.K., Marx, J.C., Neel, M.D., McNall, R.Y.,

Muul, L. and Hofmann, T. (2002). Isolated allogeneic bone marrow-derived

mesenchymal cells engraft and stimulate growth in children with

osteogenesis imperfecta: implications for cell therapy of bone. Proc Natl

Acad Sci USA., 99: 8932-8937.

Horwitz, E.M., Le Blanc, K., Dominici, M., Mueller, I., Slaper-Cortenbach, I.,

Marini, F.C., Deans, R.J., Krause, D.S. and Keating, A. (2005). Clarification

of the nomenclature for MSC: the international society for cellular therapy

position statement. Cytotherapy., 7: 393-395.

Hoste, E.A. and Kellum, J.A. (2006). Acute kidney injury: epidemiology and

diagnostic criteria. Curr Opin Crit Care., 12(6): 531-537.

Huang, J.H., Zhang, C., Zhang, D.G., Li, L., Chen, X. and Xu, D.X. (2016).

Rifampicin-induced hepatic lipid accumulation: Association with up-

regulation of peroxisome proliferator-activated receptor gamma in mouse

liver. Plos One, 11 e0165787.

Huang, D. W., Sherman, B. T. and Lempicki, R. A. (2009): “Bioinformatics

enrichment tools: paths toward the comprehensive functional analysis of

large gene lists,” Nucleic Acids Research, 37(1):1–13.

Huerta-Alardín, AL., Varon, J. and Marik, P.E. (2005). Bench-to-bedside review:

rhabdomyolysis-an overview for clinicans. Crit Care., 9(2): 158-169.

Huggins C.E, et al, (2008). Functional and metabolic remodeling in GLUT4-

deficient hearts confers hyper-responsiveness to substrate intervention.

J.Mol.Cell. Cardiol, 44:270-280.

Humphreys, B.D. and Bonventre, J.V. (2008). Mesenchymal stem cells in acute

kidney injury. Annu Rev Med.; 59: 311-325.

© COPYRIG

HT UPM

195

Humphreys, B.D., Valerius, M.T. and Kobayashi, A. et al. (2008). Intrinsic epithelial

cells repair the kidney after injury. Cell Stem Cell. 2: 284-291.

Imai, N., Kaur, T., Rosenberg, M.E. et al., (2009). Cellular therapy of kidney

diseases. Semin Dial; 22: 629–635.

Imberti, B., Morigi, M., Tomasoni, S., et al., (2007). Insulin-like growth factor-1

sustains stem cell mediated renal repair. J Am Soc Nephrol.,18: 2921-2928.

In 't Anker, P.S., Scherjon, S.A., Kleijburg-van, Dder Keur C., de Groot-Swings,

G.M., Claas, F.H., Fibbe, W.E. and Kanhai, H.H. (2004). Isolation of

mesenchymal stem cells of fetal or maternal origin from human placenta.

Stem Cells. 22: 1338-1345.

Ip, M., Cheng, K.P. and Cheung, W.C. (1991). Disseminated intravascular

coagulopathy associated with rifampicin. Tubercle., 72(4): 291-293.

Ismail, A., Fouad, O., Abdelnasser, A. et al. (2010). Stem cell therapy improves the

outcome of liver resection in cirrhotics. J Gastrointest Cancer, 41:17-23.

Isnard Bagnis, C., Deray, G., Baumelou, A., Le Quintrec, M. and Vanherweghem,

J.L. (2004). Herbs and the kidney. Am J Kidney Dis., 44(1): 1-11.

Jana C., Marie R., Hana H., Josef H., Jiri Z. and Lukas S. (2016). The mammalian

homologue of yeast Afg1 ATPase (lactation elevated 1) mediates degradation

of nuclear-encoded complex IV subunits. Biochem. J. 473:797–804.

Janeway, Charles A., Travers, Paul, Walport, Mark, Capra, J. Donald. (1999).

Immunobiology: the immune system in health and disease, 4th Edition.

London: Current Biology., 116-135.

Jiang, Y., Jahagirdar, B. N., Reinhardt, R. L. et al. (2002). Pluripotency of

mesenchymal stem cells derived from adult marrow. Nature, 418(6893):41–

49.

Jiang, H., Qu, L., Li, Y. et al., (2011). Bone marrow mesenchymal stem cells reduce

intestinal ischemia/reperfusion injuries in rats. Journal of Surgical Research,

168(1): 127–134.

Juang, Y.C.V., Tsao, T.C., Chiang, Y.C., Lin, J.L. and Tsai, Y.H. (1992). Acute

renal failure and severe thrombocytopenia induced by rifampicin: report of a

case. J Formosan Med Assoc., 91(4): 475-476.

Ju-Hyun, K., Woong, S.N., Sun, J.K., Oh, K.K., Eun, J.S., Jung, J.J., Riya S., Tae

H.L., Tae W. J., Sung H.K., Hye, S.L., and Sangkyu, L. (2017). Mechanism

Investigation of Rifampicin-Induced Liver Injury Using Comparative

Toxicoprteomics in Mice. International Journal of Molecular Sciences,

18(1417): 1-13.

© COPYRIG

HT UPM

196

Kalra, B.S., Aggarwal, S., Khurana, N. and Gupta, U. (2007). Effect of cimetidine on

hepatotoxicity induced by isoniazid-rifampicin combination in rabbits. Indian

J. Gastroenterol., 26: 18–21.

Kanathur, N., Mathai, M.G., Byrd, R.P., Fields, C.L. and Roy, T.M. (2001).

Simvastatindiltiazem drug interaction resulting in rhabdomyolysis and

hepatitis. Tenn Med., 94: 339-341.

Kaplowitz, N. (2004). Drug-induced liver injury. Clin Infect Dis., 38: S44-S48.

Kaufman, J., Dhakal, M., Patel, B. and Hamburger, R. (1991). Community-acquired

acute renal failure. Am J Kidney Dis., 17(2): 191-198.

Karaoz, E., Aksoy, A., Ayhan, S., SarIboyacI, A. E., Kaymaz, F. and Kasap, M.

(2009). Characterization of mesenchymal stem cells from rat bone marrow:

ultrastructural properties, differentiation potential and immunophenotypic

markers. Histochemistry and Cell Biology, 132 (5): 533–546.

Keating, A. (2012). Mesenchymal stromal cells: new directions. Cell Stem Cell. 10:

709–716.

Kharaziha, P., Hellstrom, P.M., Noorinayer, B., Farzaneh, F., Aghajani, K., Jafari,

F., Telkabadi, M., Atashi, A., Honardoost, M. and Zali, M.R. (2009).

Improvement of liver function in liver cirrhosis patients after autologous

mesenchymal stem cell injection: a phase I-II clinical trial. Eur J

Gastroenterol Hepatol., 21: 1199-1205.

Kido, Y., Matsson, P., and Giacomini, K. M. (2011). Profiling of a prescription drug

library for potential renal drug-drug interactions mediated by the organic

cation transporter 2. J. Med. Chem. 54, 4548–4558.

Kihm A.J., Kong Y., Hong W., Russell J.E., Rouda S., Adachi K., Simon

M.C., Blobel G.A. and Weiss M.J. (2002). An abundant erythroid protein that

stabilizes free alpha-haemoglobin. Nature 417:758-763.

Kikkawa, R., Fujikawa, M., Yamamoto, T., Hamada, Y., Yamada, H. and Horii, I.

(2006). In vivo hepatotoxicity study of rats in comparison with in vitro

hepatotoxicity screening system. J. Toxicol. Sci., 31: 23-34.

Kim, Y., Kim, H., Cho, H., Bae, Y., Suh, K. and Jung, J. (2007). Direct comparison

of human mesenchymal stem cells derived from adipose tissues and bone

marrow in mediating neovascularization in response to vascular ischemia.

Cell Physiol Biochem., 20: 867-876.

Kim, Y. N., Kim, S., Kim, I.-Y. et al. (2013). Transcriptomic analysis of

insulin-sensitive tissues from anti-diabetic drug treated ZDF rats, a

T2DM animal model, PLoS ONE, 8(7), Article ID e69624.

© COPYRIG

HT UPM

197

Kim, D., Pertea, G., Trapnell, C., Pimentel, H., Kelley, R. and Salzberg, S. L.

(2014): “TopHat2: accurate alignment of transcriptomes in the presence of

insertions, deletions and gene fusions,” Genome Biology, 14(4) article R36.

Kinnaird, T., Stabile, E., Burnett, M.S., Lee, C.W., Barr, S., Fuchs, S. and Epstein,

S.E. (2004). Marrow-derived stromal cells express genes encoding a broad

spectrum of arteriogenic cytokines and promote in vitro and in vivo

arteriogenesis through paracrine mechanisms. Circ Res., 94: 678-685.

Kinnaird, T., Stabile, E., Burnett, M.S., Shou, M., Lee, C.W., Barr, S., Fuchs, S. and

Epstein, S.E. (2004). Local delivery of marrow-derived stromal cells

augments collateral perfusion through paracrine mechanisms. Circulation.

109: 1543-1549.

Kishi, M., Emi, Y., Sakaguchi, M., Ikushiro, S., Iyanagi, T. (2008). Ontogenic

isoform switching of UDP-glucuronosyltransferase family 1 in rat liver.

Biochemical and biophysical research communications 377: 815–819.

Kleinknecht, D. and Adhemar, J.P. (1977) Les insufficances renales aigues dues a la

rifampicine. Med Mal Infect., 7: 117-119.

Kobayashi, C.I and Suda, T. (2012). Regulation of reactive oxygen species in stem

cells and cancer stem cells. J Cell Physiol., 227:421e430.

Kobayashi, K., Yoshida, A., Ejiri, Y., Takagi, S., Mimura. H. et al. (2012). Increased

expression of drug-metabolizing enzymes in human hepatocarcinoma FLC-4

cells cultured on micro-space cell culture plates. Drug metabolism and

pharmacokinetics.

Koc, O.N. and Lazarus, H.M. (2001). Mesenchymal stem cells: heading into the

clinic. Bone Marrow Transplant., 27: 235-239.

Koc, O.N., Day, J., Nieder, M., Gerson, S.L., Lazarus, H.M. and Krivit, W. (2002).

Allogeneic mesenchymal stem cell infusion for treatment of metachromatic

leukodystrophy (MLD) and hurler syndrome (MPS-IH). Bone Marrow

Transplant., 30: 215-222.

Kodner, C.M. and Kudrimoti, A. (2003). Diagnosis and management of acute

interstitial nephritis. Am Fam Physician., 67(12): 2527-2534.

Kohli, H.S., Bhaskaran, M.C., Muthukumar, T., et al. (2000). Treatment-related

acute renal failure in the elderly: a hospital-based prospective study. Nephrol

Dial Transplant., 15(2): 212-217.

Kopen, G.C., Prockop, D.J. and Phinney, D.G. (1999). Marrow stromal cells migrate

throughout forebrain and cerebellum, and they differentiate into astrocytes

after injection into neonatal mouse brains. Proc Natl Acad Sci U S A, 96:

10711-10716.

© COPYRIG

HT UPM

198

Kotton, D.N., Ma, B.Y., Cardoso, W.V., Sanderson, E.A., Summer, R.S., Williams,

M.C. and Fine, A. (2001). Bone marrow-derived cells as progenitors of lung

alveolar epithelium. Development. 128: 5181-5188.

Krampera, M., Cosmi, L., Angeli, R., Pasini, A., Liotta, F., Andreini, A., Santarlasci,

V., Mazzinghi, B., Pizzolo, G. and Vinante, F, et al. (2006). Role for

interferon-gamma in the immunomodulatory activity of human bone marrow

mesenchymal stem cells. Stem Cells., 24: 386-398.

Krasnodembskaya, A., Song, Y., Fang, X., Gupta, N., Serikov, V., Lee, J.W. and

Matthay, M.A. (2010). Antibacterial effect of human mesenchymal stem cells

is mediated in part from secretion of the antimicrobial peptide LL-37. Stem

Cells. 28: 2229-2238.

Krejcie and Morgan (1970). Determining sample size for research activities.

Educational and psychological measurement. Vol.30, pp. 607-610.

Kremer, J.M., Lee, R.G. and Tolman, K.G. (1989). Liver histology in rheumatoid

arthritis patients receiving long-term methotrexate therapy: a prospective

study with baseline and sequential biopsy samples. Arthritis Rheum., 32: 121-

127.

Kumar, A., Misra, P.K., Mehotra, R., Govil, Y.C. and Rana, G.S. (1991).

Hepatotoxicity of rifampin and isoniazid: is it all drug-induced hepatitis? Am

Rev Respir Dis., 143: 1350-1352.

Kumar, A., Toledo, J.C., Patel, R.P., Lancaster, J.R. Jr and Steyn, A.J. (2007).

Mycobacterium tuberculosis DosS is a redox sensor and DosT is a hypoxia

sensor. Proc Natl Acad Sci U S A, 104: 11568e11573.

Kumar, S. and Ponnazhagan, S. (2007). Bone homing of mesenchymal stem cells by

ectopic alpha 4 integrin expression. FASEB J; 21: 3917-3927.

Kurte, M., Bravo-Algeria, J., Torres, A., Carrasco, V., Ibanez, C., Vega-letter, A.M.,

Fernandez-O, C., Irarrazabal, C.E., Figueroa, F.E., Fuentealba, R.A., Riedel,

C. and carrion, F. (2015): Intravenous administration of bone marrow-

derived mesenchymal stem cells induced a switch from classical to atypical

symptoms in experimental autoimmune encephalomyelitis. 1-14.

http://dx.doi.org/10.1155/2015/140170.

Lai, F.M., Lai, K.N. and Chong, W.Y. (1987). Papillary necrosis associated with

rifampicin therapy. Aust NZ J Med., 17(1): 68-70.

Lam, S. P., Luk, J. M., Man, K. et al., (2010). Activation of interleukin- 6-induced

glycoprotein 130/signal transducer and activator of transcription 3 pathway in

mesenchymal stem cells enhances hepatic differentiation, proliferation, and

liver regeneration. Liver Transplantation, 16(10): 1195-1206.

© COPYRIG

HT UPM

199

Lamy, P., Cacoub, P., Deray, G., Baumelou, A. and Detilleux, M. (1989). Acute

renal failure and nephrotic syndrome caused by rifampicin: polymorphism of

the nephrotoxicity of rifampicin. Ann Med Interne (Paris)., 140(4): 323-325.

Lange, C., Togel, F., Ittrich, H. et al., (2005). Administered mesenchymal stem cells

enhance recovery from ischemia/reperfusioninduced acute renal failure in

rats. Kidney International, 68(4): 1613-1617.

Langman, G., Hall, PM. and Todd, G. (2001). Role of non-alcoholic steatohepatitis

in methotrexate-induced liver injury. J Gastroenterol Hepatol., 16: 1395-

1401.

Larrey, D. (2002). Epidemiology and individual susceptibility to adverse drug

reactions affecting the liver. Semin Liver Dis., 22: 145-155.

Lazarus, H.M., Haynesworth, S.E., Gerson, S.L., Rosenthal, N.S. and Caplan, A.I.

(1995). Ex vivo expansion and subsequent infusion of human bone marrow-

derived stromal progenitor cells (mesenchymal progenitor cells): implications

for therapeutic use. Bone Marrow Transplant. 16: 557-564.

Laws, E.R. Jr. (1991). Conservative surgery and radiation for childhood

craniopharyngiomas. Journal of neurosurgery 74: 1025–1026.

Le Blanc, K. (2003). Immunomodulatory effects of fetal and adult mesenchymal

stem cells. Cytotherapy., 5: 485-489.

Lee, W. S., Suzuki, Y., Graves, S. S. et al. (2011). Canine bone marrowderived

mesenchymal stromal cells suppress alloreactive lymphocyte proliferation in

vitro but fail to enhance engraftment in canine bone marrow transplantation.

Biology of Blood and Marrow Transplantation, 17(4):465–475.

Lee, D. S., Gil, W. H., Lee, H. H. et al., (2004). Factors affecting graft survival after

living donor liver transplantation. Transplantation Proceedings, 36(8): 2255-

2256.

Lee, H.A. (1979). Drug induced diseases, drug related disease and the kidney. Br.

Med. J., 2: 104-107.

Lee, J.W., Fang, X., Gupta, N., Serikov, V., Matthay, M.A. (2009). Allogeneic

human mesenchymal stem cells for treatment of E. coli endotoxin-induced

acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S

A, 106: 16357-16362.

Lee, R.H, Seo, M.J., Reger, R.L., Spees, J.L., Pulin, A.A., Olson, S.D. and Prockop,

D.J. (2006). Multipotent stromal cells from human marrow home to and

promote repair of pancreatic islets and renal glomeruli in diabetic NOD/scid

mice. Proc Natl Acad Sci U S A, 103: 17438-17443.

© COPYRIG

HT UPM

200

Lee, S.J., Lee, Y.S., Zimmers, T.A., Soleimani, A., Matzuk, M.M. et al. (2010).

Regulation of muscle mass by follistatin and activins. Molecular

endocrinology 24: 1998–2008.

Lefford, M.J, McGregor, D.D. and Mackaness, G.B. (1973). Immune response to

Mycobacterium tuberculosis in rats. Infect Immun., 8: 182-189.

Levine, M., Collin, K. and Kassen, B.O. (1991). Acute hemolysis and renal failure

following discontinuous use of rifampin. DICP., 25(7-8): 743-744.

Li, K., Han, Q., Yan, X., Liao, L. and Zhao, R.C. (2010): Not a process of simple

vicariousness, the differentiation of human adipose-derived mesenchymal

stem cells to renal tubular epithelial cells plays an important role in acute

kidney injury repairing. Stem Cells Dev., 19: 1267-1275.

Li, R., Xu, W., Wang, Z., Liang, B., Wu, J.-R., and Zeng, R. (2012). Proteomic

characteristics of the liver and skeletal muscle in the Chinese tree shrew

(Tupaia belangeri chinensis), Protein & Cell, 3(9): 691–700,.

Liechty, K.W., MacKenzie, T.C., Shaaban, A.F., Radu, A., Moseley, A.M., Deans,

R., Marshak, D.R. and Flake, A.W. (2000): Human mesenchymal stem cells

engraft and demonstrate site-specific differentiation after in utero

transplantation in sheep. Nat Med., 6: 1282-1286.

Lillebaek, T., Dirksen A., Vynnycky, E., Baess, I., Thomsen, V.O and Andersen,

A.B. (2003). Stability of DNA patterns and evidence of Mycobacterium

tuberculosis reactivation occurring decades after the initial infection. J Infect

Dis., 188:1032e1039.

Lin, P.L., Rodgers, M., Smith, L., Bigbee, M., Myers, A, et al. (2009). Quantitative

comparison of active and latent tuberculosis in the cynomolgus macaque

model. Infect Immun., 77(10): 4631-42.

Liao, Y., Zhang, L.S and Peng, S.Q (2011). Gene Expression Profiles of Rifampicin-

Injured Rat Liver Using cDNA Microarray. Chinese Journal of New Drugs,

20(13): 1212-1216.

Liao, Y., Zhang, L.S and Peng, S.Q (2011). Proteomic Analysis Isoniazid Toxicity in

Rat Liver. Chinese Journal of New Drugs, 20(1): 18-23.

Liao, Y., Zhang, L.S and Peng, S.Q (2015). Gene Expression Profiles in Injured Rat

Liver Induced by Isoniazid and Rifampicin Using cDNA Microarray.

Chinese Journal of New Drugs, 24(11): 1278-1284.

Liu, Y., Yan, X., Sun, Z., Chen, B., Han, Q., Li, J. and Zhao, R.C. (2007). Flk-1+

adipose-derived mesenchymal stem cells differentiate into skeletal muscle

satellite cells and ameliorate muscular dystrophy in mdx mice. Stem Cells

Dev., 16: 695-706.

© COPYRIG

HT UPM

201

Londos, C., Brasaemle, D.L., Schultz, C.J., Segrest, J.P., Kimmel, A.R. (1999).

Perilipins, ADRP, and other proteins that associate with intracellular neutral

lipid droplets in animal cells. Semin. Cell Dev. Biol., 10: 51–58.

Lonnroth, K and Raviglione, M. (2008). Global epidemiology of tuberculosis:

prospects for control. Semin Respir Crit Care Med., 29: 481e491.

McCabe, M., Waters, S., Morris, D., Kenny, D., Lynn, D. and Creevey, C. (2012).

RNA-seq analysis of differential gene expression in liver from lactating dairy

cows divergent in negative energy balance, BMC Genomics, 13(193).

M’iasnykov, V.H. (1993). Acute hepatorenal failure occurring after taking

rifampicin. Vrach Delo., (7): 124-126.

Machado, A.L.D., Branda˜o, A.A.H., da Silva, C.M.O.M. and da Rocha, R.F.

(2003). Influence of tetracycline in the hepatic and renal development of rat’s

offspring. Braz. Arch. Biol. Technol. Int. J., 46(1): 47-51.

Manescu, N., Gluhovschi, G., Golea, O., Nicolcioiu, M., Schwarzkopf, A. and Zosin,

C. (1974). Akutes Nierenversagen nach Rifampicm. Munch Med

Wochenschr; 116: 2161-2163.

Manor, S.M., Guillory, G.S. and Jain, S.P. (2004). Clopidogrel-induced thrombotic

thrombocytopenic purpura-hemolytic uremic syndrome after coronary artery

stenting. Pharmacotherapy., 24(5): 664-667.

Marde Arrhen, Y., Nylen, H., Lovgren-Sandblom, A., Kanebratt, K.P., Wide, K. and

Diczfalusy, U. A. (2013). Comparison of 4β-hydroxycholesterol: Cholesterol

and 6β-hydroxycortisol: Cortisol as markers of CYP3A4 induction. Br. J.

Clin. Pharmacol., 75: 1536–1540.

Marina M. B., Danilo P., Daniela B., Marilena C., Stefania P., Cinzia B., Martina C.,

Rachele Del S., Francesca F., Angelo S., Paolo P., Luigina R., Giuseppe S.,

and Maria A. D. F. (2014). NEDD4 controls the expression of GUCD1, a

protein upregulated in proliferating liver cells. Cell Cycle 13:(12)1902–1911.

Markowitz, G.S. and Perazella, M.A. (2005). Drug-induced renal failure: a focus on

tubulointerstitial disease. Clin Chim Acta., 351(1-2): 31-47.

Markowitz, G.S., Appel, G.B., Fine, P.L., et al. (2001). Collapsing focal segmental

glomerulosclerosis following treatment with high-dose pamidronate. J Am

Soc Nephrol., 12(6): 1164-1172.

Markowitz, G.S., Fine, P.L., Stack, J.I., et al. (2003). Toxic acute tubular necrosis

following treatment with zoledronate (Zometa). Kidney Int., 64(1): 281-289.

Martin, C.M., Ferdous, A., Gallardo, T., Humphries, C., Sadek, H., Caprioli, A.,

Garcia, J.A., Szweda, L.I., Garry, M.G and Garry, D.J (2008).

Hypoxiainducible factor-2a transactivates Abcg2 and promotes

cytoprotection in cardiac side population cells. Circ Res., 102:1075e1081.

© COPYRIG

HT UPM

202

Martin-Carbonero, L., Nunez, M., Gonzalez-Lahoz, J. and Soriano, V. (2003).

Incidence of liver injury after beginning antiretroviral therapy with efavirenz

or nevirapine. HIV Clin Trials., 4: 115-120.

Martinez, E., Collazos, J. and Mayo, J. (1999). Hypersensitivity reactions to

rifampin. Medicine, 78: 361-369.

Marzouk, M., Hassaneen, S., Hussein, H. and Sammaan, H.A. (2009). Ameliorative

effect of Mepacure against rimactazid-induced hepatotoxicity in rats. Aust. J.

Basic Appl. Sci., 3(3): 2350-2354.

Mathur, A. and Martin, J.F. (2004). Stem cells and repair of the heart. Lancet,

365(9429): 183-192.

Mauri, J.M., Fort, J., Bartolome, J., et al. (1982). Antirifampicin antibodies in acute

rifampicin-associated renal failure. Nephron., 31: 177-179.

McMurray, D.N. (1994). Guinea pig model of tuberculosis. In: Bloom BR, editor.

Tuberculosis: pathogenesis, protection and control. Washington, SC: ASM

Press. pp. 135-147.

McGarry, J. D., Mannaerts, G. P. and Foster, D. W. (1977). A possible role for

malonyl-CoA in the regulation of hepatic fatty acid oxidation and

ketogenesis, The Journal of Clinical Investigation, 60(1):265–270.

Mediavilla Garcia, J.D., Lopez-Gomez, M., Arrebola Nacle, J.P., Oyonarte, S. and

Jimenez-Alonso, J. (1992). Hemolytic anemia and acute renal failure caused

by rifampicin. Anales de Medicina Interna., 9(5): 259.

Meirelles, L.S and Nardi, N.B. (2003). Murine marrow-derived mesenchymal stem

cell: isolation, in vitro expansion, and characterization. British Journal of

Heamatology, 123:702-711.

Meleady, P and O’Connor, R. (2006). General Procedures for Cell Culture. In Cell

Biology, A laboratory Handbook (3rd Ed., Vol. 1), J.Celis, eds. (Elsevier), pp.

13-20.

Menzies, D., Al Jahdali, H and Al Otaibi, B. (2011). Recent developments in

treatment of latent tuberculosis infection. Indian J Med Res, 133: 257e266.

Menzies, D., Dion, M.J., Rabinovitch, B., Mannix, S., Brassard, P. and

Schwartzman, K. (2004). Treatment completion and costs of a randomized

trial of rifampin for 4 months versus isoniazid for 9 months. Am J Respir Crit

Care Med., 170: 445-449.

Merrick, B.A., Phadke, P.D., Auerbach, S.S., Mav, D., Stiegelmeyer, M.S., Shah,

R.S. and Tice, R.R. (2013): RNA-Seq profiling reveals novel hepatic gene

expression pattern in aflatoxin B1 treated rats. Plos one, 8(4): 1-19.

© COPYRIG

HT UPM

203

Mesnage, R., Matthew, A., Manuela, C., Manuela, M., Gilles-Eric, S and Michael,

N. A. (2015). Transcriptome profile analysis reflects rat liver and kidney

damage following chronic ultra-low dose Roundup exposure. Environmental

Health, 14:70

Michalopoulos, G.K. (2010). Liver regeneration after partial hepatectomy. Critical

analysis of mechanistic dilemmas. Am J Pathol., 176:2-13.

Miller, G. and McGarity, G.J. (2009). Tetracycline-induced renal failure after dental

treatment. JADA., 140(1): 56-60.

Miyata, N., Taniguchi, K., Seki, T., Ishimoto, T., Sato-Watanabe, M., Yasuda, Y.,

Doi, M., Kametani, S., Tomishima, Y., Ueki, T. et al. (2001). Het0016, a

potent and selective inhibitor of 20-HETE synthesizing enzyme. Br. J.

Pharmacol., 133: 325–329.

Miyajima, A., Kinoshita, T., Tanaka, M., Kamiya, A., Mukouyama, Y. and Hara, T.

(2000). Role of oncostatin M in hematopoiesis and liver development.

Cytokine and Growth Factor Reviews, 11(3): 177-183.

Mohamadnejad, M., Alimoghaddam, K., Mohyeddin-Bonab, M., Bagheri, M.,

Bashtar, M., Ghanaati, H., Baharvand, H., Ghavamzadeh, A. and

Malekzadeh, R. (2007). Phase 1 trial of autologous bone marrow

mesenchymal stem cell transplantation in patients with decompensated liver

cirrhosis. Arch Iran Med., 10: 459-466.

Morigi, M., Imberti, B., Zoja, C. et al., (2004). Mesenchymal stem cells are

renotropic, helping to repair the kidney and improve function in acute renal

failure. Journal of the American Society of Nephrology, 15(7): 1794-1804.

Murray, A.N., Cassidy, M.J. and Templecamp, C. (1987). Rapidly progressive

glomerulonephritis associated with rifampicin therapy for pul monary

tuberculosis. Nephron, 46(4): 373-376.

Muthukumar, T., Jayakumar, M., Fernando, E.M. and Muthusethupthi, M.A. (2002).

Acute renal failure due to rifampicin. A study of 25 patients. Am. J. Kidney

Dis., 40: 690-696.

Nash, K., Hafeez, A. and Hou, S. (2002). Hospital-acquired renal insufficiency. Am J

Kidney Dis., 39(5): 930-936.

Nayak, S., Sashidhar, R.B. (2010). Metabolic intervention of aflatoxin B1 toxicity by

curcumin. Journal of ethnopharmacology 127: 641–644.

Nemeth, K., Leelahavanichkul, A., Yuen, P.S., Mayer, B., Parmelee, A., Doi, K.,

Robey, P.G., Leelahavanichkul, K., Koller, B.H. and Brown, J.M. (2009).

Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent

reprogramming of host macrophages to increase their interleukin-10

production. Nat. Med., 15: 42-49.

© COPYRIG

HT UPM

204

Nessi, H., Bonoldi, G.L., Redaelli, B. and Di Fillipo, G. (1976). Acute renal failure

after rifampicin: a case report and survey of the literature. Nephron., 16: 148–

153.

Neugarten, J., Gallo, G.R. and Baldwin, D.S. (1983). Rifampicin induced nephrotic

syndrome and acute interstitial nephritis. Am J Nephrol., 3: 38-40.

Niemi, M., Backman, J.T, Fromm, M.F., Neuvonen, P.J. and Kivisto, K.T. (2003).

Pharmacokinetic interactions with rifampicin: clinical relevance. Clin

Pharmacokinet., 42:819-850.

Noort, W. A., Oerlemans, M. I., Rozemuller, H. et al., (2012). Human versus porcine

mesenchymal stromal cells: phenotype, differentiation potential,

immunomodulation and cardiac improvement after transplantation. Journal

of Cellular and Molecular Medicine. In press.

Olyaei, A.J., de Mattos, A.M. and Bennett, W.M. (1999). Immunosuppressant-

induced nephropathy: pathophysiology, incidence and management. Drug

Saf., 21(6): 471-488.

Ornstein, G.B. and Steinbach, M.M. (1925). The resistance of the albino rat to

infection with tubercle bacilli. Am. Rev Tuberc., 12: 77-86.

Ortiz, L. A., DuTreil, M., Fattman, C. et al., (2007). Interleukin 1 receptor antagonist

mediates the anti-inflammatory and antifibrotic effect of mesenchymal stem

cells during lung injury. Proceedings of the National Academy of Sciences of

the United States of America, 104(26): 11002-11007.

Ortiz, L.A., Dutreil, M., Fattman, C., Pandey, A.C., Torres, G., Go, K. and Phinney,

D.G. (2007). Interleukin 1 receptor antagonist mediates the antiinflammatory

and antifibrotic effect of mesenchymal stem cells during lung injury. Proc

Natl Acad Sci U S A. 104: 11002-11007.

Ortiz, L.A., Gambelli, F., McBride, C., Gaupp, D., Baddoo, M., Kaminski, N. and

Phinney, D.G. (2003). Mesenchymal stem cell engraftment in lung is

enhanced in response to bleomycin exposure and ameliorates its fibrotic

effects. Proc Natl Acad Sci U S A, 100: 8407-8411.

Ostapowicz, G.M., Fontana, R., Schiødt, F., Larson, A., Davern, T., Steven Han, H.,

McCashland, T., Shakil, A., Hay, J., Hynan L, et al. (2002). Results of a

prospective study of acute liver failure at 17 tertiary care centers in the

United States. Ann Intern Med., 137: 947-954.

Otto, W.R. and Wright, N.A. (2011). Mesenchymal stem cells: from experiment to

clinic. Fibrogenesis Tissue Repair., 4: 20-10.1186/1755-1536-4-20.

Ooe, H., Chen, Q., Kon, J., Sasaki, K., Miyoshi, H. et al. (2012). Proliferation of rat

small hepatocytes requires follistatin expression. Journal of cellular

physiology 227: 2363–2370.

© COPYRIG

HT UPM

205

Pal, R., Rana, S.V., Vaiphei, K. and Singh K. (2008). Isoniazid-rifampicin induced

lipid changes in rats. Clin. Chem. Acta, 389: 55-60.

Palmer, B.F. (2002). Renal dysfunction complicating the treatment of hypertension.

N Engl J Med., 347(16): 1256-1261.

Papastavros, T., Dolovich, L.R., Holbrook, A.,Whitehead, L. and Loeb, M. (2002).

Adverse events associated with pyrazinamide and levofloxacin in the

treatment of latent multidrug-resistant tuberculosis. CMAJ., 167: 131-136.

Patternson T.A, et al. (2006). Performance comparison of one-color and two color

platforms within the micro-array quality control (MAQC) project. Nat.

Biotechnol, 24:1140-1150.

Peart, M.J, et al. (2011). Identification and Functional significance of genes

regulated by structurally different histone deacatylase inhibitors. Proc. Natl.

Acad. Sci, USA. 102:3697-3702.

Parekkadan, B., van Poll, D., Suganuma, K., Carter, E.A., Berthiaume, F., Tilles,

A.W. and Yarmush, M.L. (2007). Mesenchymal stem cell-derived molecules

reverse fulminant hepatic failure. PLoS One. 2: e941-10.1371.

Parmar, K., Mauch, P., Vergilio, J.A., Sackstein, R and Down, J.D. (2007).

Distribution of hematopoietic stem cells in the bone marrow according to

regional hypoxia. Proc Natl Acad Sci U S A, 104:5431e5436.

Pittenger, M. F., Mackay, A. M., Beck, S. C. et al., (1999). Multilineage potential of

adult human mesenchymal stem cells. Science, 284(5411): 143–147.

Peister, A., Mellad, J. A., Larson, B. L., Hall, B. M., Gibson, L. F. and Prockop, D.

J. (2004): Adult stem cells from bone marrow (MSCs) isolated from different

strains of inbred mice vary in surface epitopes, rates of proliferation, and

differentiation potential. Blood, 103(5):1662–1668.

Pelaez, E., Rodriguez, J.C., Cigarran, S. and Pereira, A. (1993). Acute renal failure

caused by two single doses of rifampicin with a year of interval. Nephron.,

64(1): 152-153.

Pelekanos, R. A., Li, J., Gongora, M. et al. (2012). Comprehensive transcriptome

and immunophenotype analysis of renal and cardiac MSC-like populations

supports strong congruence with bone marrow MSC despite maintenance of

distinct identities. Stem Cell Research, 8(1):58–73.

Peng, L., Xie, D.Y., Lin, B.L. et al. (2011). Autologous bone marrow mesenchymal

stem cell transplantation in liver failure patients caused by hepatitis B: short-

term and long-term outcomes.

Hepatology. http://dx.doi.org/10.1002/hep.24434.

Perazella, M.A. (1999). Crystal-induced acute renal failure. Am J Med., 106(4): 459-

465.

© COPYRIG

HT UPM

206

Perazella, M.A. (2005). Drug-induced nephropathy: an update. Expert Opin Drug

Saf., 4(4): 689-706.

Pereira, A., Sanz, C., Cervantes, F. and Castillo, R. (1991). Immune hemolytic

anemia and renal failure associated with rifampicin-dependent antibodies

with anti-I specificity. Ann Hematol., 63(1): 56-58.

Perico, N., Casiraghi, F., Introna, M. et al. (2011). Autologous mesenchymal

stromal cells and kidney transplantation: a pilot study of safety and clinical

feasibility. Clin J Am Soc Nephrol., 6: 412-422.

Perico, N., Casiraghi, F., Introna, M., Gotti, E., Todeschini, M., Cavinato, R.A.,

Capelli, C., Rambaldi, A., Cassis, P., Rizzo, P., Cortinovis, M., Marasà, M.,

Golay, J., Noris, M. and Remuzzi, G. (2011). Autologous mesenchymal

stromal cells and kidney transplantation: a pilot study of safety and clinical

feasibility. Clin J Am Soc Nephrol., 6: 412-422.

Perneger, T.V, Whelton, P.K. and Klag, M.J. (1994). Risk of kidney failure

associated with the use of acetaminophen, aspirin, and nonsteroidal

antiinflammatory drugs. N Engl J Med., 331(25): 1675-1679.

Philippe, H., Maryse, Fauville-Dufaux., Didier, B., Bernadette, Van V., Katia, P.,

Chuong, D., Thi, M., Sneyers, M.W., René S. and William, M. (2006).

Biosafety Recommendations for the Contained Use of Mycobacterium

tuberculosis Complex Isolates in Industrialized Countries. Royal Library of

Belgium Deposit Number D//2505/22.

Pinzani, M., Romanelli, R.G. and Magli, S. (2001). Progression of fibrosis in chronic

liver diseases – time to tally the score. J Hepatol., 34: 764-767.

Piscaglia, A.C., Campanale, R., Gasbarrini, G. et al. (2010). Stem cell-based

therapies for liver diseases: state of the art and new perspectives. Stem Cells

Int.

Pisoni, R., Ruggenenti, P. and Remuzzi, G. (2001). Drug-induced thrombotic

microangiopathy: incidence, prevention and management. Drug Saf., 24(7):

491-501.

Plumb, D.C. (1999): Veterinary Drug Handbook, third ed. Iowa State University

Press/Ames, Minnesota, pp. 654-656.

Podolsky, D. K. (2002). Inflammatory bowel disease. The New England Journal of

Medicine, 347(6): 417-429.

Poole, G., Stradling, P. and Worledge, S. (1971). Potential serious side effects of

high-dose twice weekly rifampicin. Br Med J., 3: 343.

© COPYRIG

HT UPM

207

Popp, F.C., Eggenhofer, E., Renner, P., Slowik, P., Lang, S.A., Kaspar, H., Geissler,

E.K., Piso, P., Schlitt, H.J. and Dahlke, M.H. (2008). Mesenchymal stem

cells can induce long-term acceptance of solid organ allografts in synergy

with low-dose mycophenolate. Transpl. Immunol., 20: 55-60.

Porter, R.M., Huckle, W.R. and Goldstein, A.S. (2003). Effect of dexamethasone

withdrawal on osteoblastic differentiation of bone marrow stromal cells. J

cell Biochem 90:13-22.

Power, D.A., Russel, G., Smith, F.W., et al. (1983). Acute renal failure due to

continuous rifampicin. Clin Nephrol., 20: 155-156.

Prendergast, B.D. and George, C.F. (1993). Drug-induced rhabdomyolysis—

mechanisms and management. Postgrad Med J., 69(811): 333-336.

Prince, M.I., Burt, A.D. and Jones, D.E. (2002). Hepatitis and liver dysfunction with

rifampicin therapy for pruritus in primary biliary cirrhosis. Gut, 50: 436-439.

Prockop, D.J. (1997): Marrow stromal cells as stem cells for nonhematopoietic

tissues. Science; 276:71–74.

Quevedo, H. C., Hatzistergos, K. E., Oskouei, B. N. et al.,( 2009). Allogeneic

mesenchymal stem cells restore cardiac function in chronic ischemic

cardiomyopathy via trilineage differentiating capacity. Proceedings of the

National Academy of Sciences of the United States of America, 106(33):

14022-14027.

Quinn, B.P. and Wall, B.M. (1989): Nephrogenic diabetes insipidus and tubulo-

interstitial nephritis during continuous therapy with rifampin. Am J

KidneyDis., 14(3): 217-220.

Qunibi, W.Y., Godwin, J. and Eknoyan, G. (1980). Toxic nephropathy during

continuous rifampin therapy. South Med J., 73: 791-793.

Rae, J.M., Johnson, M.D., Lippman, M.E. and Flockhart, D.A. (2001). Rifampin is a

selective, pleiotropic inducer of drug metabolism genes in human

hepatocytes: studies with cDNA and oligonucleotide expression arrays. J

Pharmacol Exp Ther., 299:849-857.

Rafiq, S., Iqbal, T., Jamil, A. and Khan, F.H. (2010). Pharmacokinetic studies of

rifampicin in healthy volunteers and tuberculosis patients. Int. J. Agric. Biol.,

12(3): 391-395.

Raouf, A. et al. (2008). Transcriptome analysis of the normal human mammary cell

commitment and differentiation process. Cell Stem Cell. 3:109-118.

Rajan, M.A., Soundararajan, R., Krishnamuthy, V. and Ramu, G. (1987). Acute

renal failure following rifampicin. Indian J Lepr; 59(3): 286-292.

© COPYRIG

HT UPM

208

Ramsopal, V., Leonard, C. and Bhathena, D. (1973). Acute renal failure associated

with rifampicin. Lancet; 1: 1195.

Ramsköld, D., Wang, E. T., Burge, C. B. and Sandberg, R. (2009). An abundance of

ubiquitously expressed genes revealed by tissue transcriptome sequence

data, PLoS Computational Biology, 5(12) Article ID e1000598.

Ramayo-Caldas, Y., Mach, N., Esteve-Codina, A. et al. (2012). Liver transcriptome

profile in pigs with extreme phenotypes of intramuscular fatty acid

composition, BMC Genomics, 13(547).

Rana, S.V., Pal, R., Vaiphei, K., Ola, R.P. and Singh, K. (2010). Hepatoprotection

by carotenoids in isoniazid- rifampicin induced hepatic injury in rats.

Biochem. Cell Biol. NBC Res. Press, 88(5): 819-834.

Rani, V.P. and Meera, K.A. (2014): Isolation and immunophenotypying

characterization of bone marrow derived stem cells. International journal of

pharmaceutical science invention, 3(4):26-32.

Rappolee, D. A., Basilico, C., Patel, Y. and Werb, Z. (1994). Expression and

function of FGF-4 in peri-implantation development in mouse embryos.

Development, 120(8): 2259-2269.

Reinders, M.E., de Fijter, J.W., Roelofs, H., Bajema, I.M., de Vries, D.K.,

Schaapherder, A.F., Claas, F.H., van Miert, P.P., Roelen, D.L., van Kooten,

C., Fibbe, W.E. and Rabelink, T.J. (2013). Autologous bone marrow-derived

mesenchymal stromal cells for the treatment of allograft rejection after renal

transplantation: results of a phase I study. Stem Cells Transl Med., 2: 107-

111.

Reinders, MEJ, de Fijter, J.W., Roelofs, H. et al. (2013). Autologous bone

marrowderived mesenchymal stromal cells for the treatment of allograft

rejection after renal transplantation: results of a phase I study. Stem Cells

Transl Med., 2: 107-111.

Reinhold, W.C., Erliandri, I., Liu, H., Zoppoli, G., Pommier, Y. et al. (2011).

Identification of a predominant co-regulation among kinetochore genes,

prospective regulatory elements, and association with genomic instability.

PloS one 6: e25991.

Rekha, V.V., Santha, T and Jawahar, M.S. (2005). Rifampicin induced renal toxicity

during retreatment of patients with pulmonary tuberculosis. J Assoc

Physician India., 53: 811-3.

Ridzuan, N., Al Abbar, A., Yip, W.K., Maqbool, M and Rajesh, R. (2016).

Characterization and expression of senescence markers in prolonged

passages of rat bone marrow-derived mesenchymal stem cells. Stem cells

international, 1-14. http://dx.doi.org/10.1155/2016/8487264

© COPYRIG

HT UPM

209

Ridzon, R., Meador, J., Maxwell, R., Higgins, K., Weismuller, P. and Onorato, I.M.

(1997). Asymptomatic hepatitis in persons who received alternative

preventive therapy with pyrazinamide and ofloxacin. Clin Infect Dis., 24:

1264-1265.

Riska, H., Matsson, K., Kock, B., et al. (1976). Acute renal failure subsequent to the

administration of rifampicin. A follow-up study of cases reported earlier.

Scand J Resp Dis., 57: 183-187.

Rojas, M., Xu, J., Woods, C.R., Mora, A.L., Spears, W., Roman, J. and Brigham,

K.L. (2005). Bone marrow-derived mesenchymal stem cells in repair of the

injured lung. Am J Respir Cell Mol Biol., 33: 145-152.

Rossert, J. (2001). Drug-induced acute interstitial nephritis. Kidney Int.; 60(2): 804-

817.

Rozemuller, H., Prins, H. J., Naaijkens, B., Staal, J., Buhring, H. J. and Martens, A.

C. (2010). Prospective isolation of mesenchymal stem cells from multiple

mammalian species using cross-reacting anti-human monoclonal antibodies.

Stem Cells and Development, 19 (12):1911–1921.

Saadi, G., Fadel, F., Ansary, E.l. M. et al., (2013). Mesenchymal stem cell

transfusion for desensitization of positive lymphocyte cross-match before

kidney transplantation: outcome of 3 cases. Cell Prolif., 46: 121-126.

Sacerdoti, D.; Balazy, M.; Angeli, P.; Gatta, A.; McGiff, J.C. Eicosanoid excretion

in hepatic cirrhosis. Predominance of 20-HETE. J. Clin. Investig. 1997, 100,

1264.

Salem, H.K. and Thiemermann, C. (2010). Mesenchymal stromal cells: current

understanding and clinical status. Stem Cells. 28: 585-596.

Salih, S.B., Kharal, M., Qahtani, M., Dahneem, L. and Nohair, S. (2008). Acute

interstitial nephritis induced by intermittent use of rifampicin in patient with

brucellosis. Saudi J. Kidney Dis. Transpl. 19(3): 450-452.

Samuel, S., Kayode, O., Musa, O. I., Nwigwe, G.C., Aboderin, A.O., Salami, T.

A.T. and Taiwo, S.S. (2011). Nosocomical infection and the challenges of

control in developing countries. Afr. J. CLN. Exper. Microbiol. (2): 102-

110.

Santhosh, S., Sini, T.K., Anandan, R. and Mathew, P.T. (2006). Effect of chitosan

supplementation on antitubercular drugs-induced hepatotoxicity in rats.

Toxicology, 219: 53-59.

Santhosh, S., Sini, T.K., Anandan, R. and Mathew, P.T. (2007). Hepatoprotective

activity of chitosan against isoniazid and rifampicin-induced toxicity in

experimental rats. Eur. J. Pharmacol., 572: 69-73.

© COPYRIG

HT UPM

210

Schetz, M., Dasta, J., Goldstein, S. and Golper, T. (2005). Drug-induced acute

kidney injury. Curr Opin Crit Care., 11(6): 555-565.

Scheuer, P.J., Lal, S., Summerfield, J.A. and Sherlock, S. (1974). Rifampicin

hepatitis. Lancet, 303(7855): 421-425.

Scheuer, P.J., Summerfield, J.A., Lal, S. and Sherlock, S. (1974). Rifampicin

hepatitis: a clinical and histological study. Lancet, 1: 421-425.

Schmidt, C., Bladt, F., Goedecke, S. et al., (1995). Scatter factor/ hepatocyte growth

factor is essential for liver development. Nature, 373(6516): 699-702.

Schnellmann, R.G., Kelly, K.J. (1999). Pathophysiology of nephrotoxic acute renal

failure. In: Berl T, Bonventre JV, eds. Acute Renal Failure. Philadel-phia,

Pa.: Blackwell Science. Schrier RW, ed. Atlas of Diseases of the Kidney, vol

1. http://www.kidneyatlas.org/book1/adk1_15.pdf. Accessed November 8,

2007.

Schoolwerth, A.C., Sica, D.A., Ballermann, B.J. and Wilcox, C.S. (2001). Renal

considerations in angiotensin converting enzyme inhibitor therapy: a

statement for healthcare professionals from the Council on the Kidney in

Cardiovascular Disease and the Council for High Blood Pressure Research of

the American Heart Association. Circulation., 104(16): 1985-1991.

Schuetz, E.G., Schinkel, A.H., Relling, M.V. and Schuetz, J.D. (1996). P-

glycoprotein: a major determinant of rifampicin-inducible expression of

cytochrome P4503A in mice and humans. Proc Natl Acad Sci USA, 93:4001-

4005.

Schuleri, K. H., Amado, L. C. Boyle, A. J. et al., (2008). Early improvement in

cardiac tissue perfusion due to mesenchymal stem cells. American Journal of

Physiology, 294(5): H2002-H2011.

Selmani, Z., Naji, A., Zidi, I., Favier, B., Gaiffe, E., Obert, L., Borg, C., Saas, P.,

Tiberghien, P. and Rouas-Freiss, N. (2008). Human leukocyte antigen-G5

secretion by human mesenchymal stem cells is required to suppress T

lymphocyte and natural killer function and to induce

CD4 + CD25highFOXP3+ regulatory T cells. Stem Cells. 26: 212-222.

Selsted ME, White SH, Wimley WC (1995). Structure, function, and membrane

integration of defensins. Curr. Opin. Struct. Biol. 5 (4): 521–5270.

Shabana, M.B., Hania, M., Soheir E.M. Khadre and Marwa G. Elemam (2012):

Influence of rifampicin and tetracycline administration on some biochemical

and histological parameters in albino rats. The journal of basic and applied

zoology. 65(5):299-308.

Shah, B.V., Almeida, A.F., Khanna, U.B. and Acharya, V.N. (1986). Rifampicin

induced acute renal failure. J Assoc Physicians India, 34(2): 159-160.

© COPYRIG

HT UPM

211

Shirley, H.J.M., Jack J.Haitsman., Claudia C.D.S., Yupu, D., Patrick F.H.L., Arthur,

S.S., Conrad, W.L and Duncan, J.S. (2010). Mesenchymal stem cells reduce

inflammation while enhancing bacterial clearance and improving survival in

sepsis. American Journal of Respiratory and Critical Care Medicine,

182(8):1047-1057.

Shu, Y., Bello, C. L., Mangravite, L. M., Feng, B., and Giacomini, K. M. (2001).

Functional characteristics and steroid hormone-mediated regulation of an

organic cation transporter in Madin-Darby canine kidney cells. J. Pharmacol.

Exp. Ther. 299, 392–398.

Silins, I., Stenius, U. and Hogberg, J. (2004). Induction of preneoplastic rat liver

lesions with an attenuated p53 response by low doses of diethylnitrosamine.

Archives of toxicology 78: 540–548.

Simpson, I.J., Marshall, M.R., Pilmore, H., et al. (2006). Proton pump inhibitors and

acute interstitial nephritis: report and analysis of 15 cases. Nephrology

(Carlton)., 11(5): 381-385.

Sipe, J.D. (2002). Tissue engineering and reparative medicine. Ann New York Acad

Sci., 961: 1-9.

Sokal, E.M., Smets, F., Bourgois, A. et al. (2003). Hepatocyte transplantation in a 4-

year-old girl with peroxisomal biogenesis disease: technique, safety, and

metabolic follow-up. Transplantation. 76: 735-738.

Sotiropoulou, P.A., Perez, S.A., Gritzapis, A.D., Baxevanis, C.N. and Papamichail,

M. (2006). Interactions between human mesenchymal stem cells and natural

killer cells. Stem Cells. 24: 74-85.

Sun, S., Guo, Z., Xiao, X. et al. (2003). Isolation of mouse marrow mesenchymal

progenitors by a novel and reliable method. Stem Cells, 21(5):527–535.

Spaeth, E., Klopp, A., Dembinski, J., Andreeff, M. and Marini, F. (2008).

Inflammation and tumor microenvironments: defining the migratory itinerary

of mesenchymal stem cells. Gene Ther., 15: 730-738.

Spaggiari, G.M., Capobianco, A., Becchetti, S., Mingari, M.C. and Moretta, L.

(2006). Mesenchymal stem cell-natural killer cell interactions: evidence that

activated NK cells are capable of killing MSCs, whereas MSCs can inhibit

IL-2-induced NK-cell proliferation. Blood. 107: 1484-1490.

Steck E., Boeuf S., Gabler J., Werth N., Schnatzer P. and Diederichs S. (2012).

Regulation of H19 and its encoded microRNA-675 in osteoarthritis and

under anabolic and catabolic in vitro conditions. J Mol Med (Berl). 90 (10):

1185–95.

Straub, B.K., Stoeffel, P., Heid, H., Zimbelmann, R. and Schirmacher, P. (2008).

Differential pattern of lipid droplet-associated proteins and de novo perilipin

expression in hepatocyte steatogenesis. Hepatology, 47: 1936–1946.

© COPYRIG

HT UPM

212

Stenberg, D. (1976). Interconcertible enzymes in adipose tissue regulated by cyclic

AMP-dependent protein kinase. Adv. Cyclic Nucleotide Res., 7: 157-198.

Stone, W.J., Waldron, J.H.D., Primm, R.D., et al., (1976). Acute diffuse interstitial

nephritis related to chemotherapy of tuberculosis. Antimicrob Agents

Chemother.; 10: 164-166.

Suh, Y. H., Kim, Y., Bang, J. H. et al. (2005). Analysis of gene expression profiles

in insulin-sensitive tissues from pre-diabetic and diabetic Zucker diabetic

fatty rats, Journal of Molecular Endocrinology, 34(2):299–315.

Suda, T., Takubo, K and Semenza, G.L. (2011). Metabolic regulation of

hematopoietic stem cells in the hypoxic niche Cell. Stem Cell, 9: 298-310.

Sugawara, I., Yamada, H. and Mizuno, S. (2004). Pulmonary tuberculosis in

spontaneously diabetic goto kakizaki rats. Tohoku J Exp Med., 204: 135-145.

Sugawara, I., Yamada, H. and Mizuno, S. (2006). Nude rat (F344/N-rnu)

tuberculosis. Cell Microbiol., 8: 661-667.

Sugawara, I., Yamada, H., Mizuno, S. (2004). Pathological and immunological

profiles of rat tuberculosis. Int J Exp Pathol., 85: 125-134.

Sullivan, M.O., Gordon-Evans, J.W., Fredericks, P.L., Kiefer, K., Conzemius, M.G.

and Griffon, D.J. (2016): Comparison of mesenchymal stem cell surface

markers from bone marrow aspirates and adipose stromal vascular fraction

sites. Front. Vet. Sci. 2:82.

Sylvester, J.E., Fischel-Ghodsian, N., Mougey, E.B and O’Brien, T.W. (2004):

Mitochondrial ribosomal proteins: Candidate genes for mitochondrial

disease. Genet Med 6(2):73–80.

Tada, T., Ohara, A., Nagai, Y., Otani, M., Ger, Y.C. and Kawamura, S. (1995). A

case report of nephrotic syndrome associated with rifampicin therapy.

Nippon Jinzo Gakkai Shi., 37(2): 145-150.

Tahan, S.R., Diamond, J.R., Blank, J.M. and Horan, R.F. (1985). Acute hemolysis

and renal failure with rifampicin-dependent antibodies after discontinuous

administration. Transfusion., 25(2): 124-127.

Takahashi, M., Li, T.S., Suzuki, R., Kobayashi, T., Ito, H., Ikeda, Y., Matsuzaki, M.

and Hamano, K. (2006). Cytokines produced by bone marrow cells can

contribute to functional improvement of the infarcted heart by protecting

cardiomyocytes from ischemic injury. Am. J Physiol Heart Circ Physiol.,

291: H886-893.

Tan, J., Wu, W., Xu, X., Liao, L., Zheng, F., Messinger, S., Sun, X., Chen, J., Yang,

S., Cai, J., Gao, X., Pileggi, A. and Ricordi, C. (2012). Induction therapy

with autologous mesenchymal stem cells in living-related kidney transplants:

a randomized controlled trial. JAMA., 307: 1169-1177.

© COPYRIG

HT UPM

213

Tan, J.,Wu, W., Xu, X. et al. (2012). Induction therapy with autologous

mesenchymal stem cells in living-related kidney transplants: a randomized

controlled trial. JAMA., 307: 1169-1177.

Tanaka, E.M. (2003). Regeneration: if they can do it, why can’t we? Cell. 113: 559-

562.

Tasduq, S.A., Peerzada, K., Koul, S., Bhat, R. and Johri, R.K. (2005). Biochemical

manifestations of anti-tuberculosis drugs induced hepatotoxicity and the

effect of Silymarin. Heptol, Res., 31: 132-135.

Tasduq, S.A., Kaiser, P., Sharma, S.C. and Johri, R.K. (2007). Potentiation of

isoniazid-induced liver toxicity by rifampicin in a combinational therapy of

antitubercular drugs (rifampicin, isoniazid and pyrazinamide) in Wistar rats:

A toxicity profile study. Hepatol. Res. 37:845-853.

Terai, S., Ishikawa, T., Omori, K. et al. (2006). Improved liver function in patients

with liver cirrhosis after autologous bone marrow cell infusion therapy. Stem

Cells. 24: 2292-2298.

Tiffin, N., Adie, E., Turner, F. et al., (2006). Computational disease gene

identification: a concert of methods prioritizes type 2 diabetes and obesity

candidate genes,” Nucleic Acids Research, 34(10):3067–3081.

Thorgeirsson, S.S. (1996). Hepatic stem cells in liver regeneration. FASEB J.,

10:1249-1256.

To¨gel, F.E. and Westenfelder, C. (2012): Kidney protection and regeneration

following acute injury: progress through stem cell therapy. Am J Kidney Dis.;

60: 1012-1022.

Togel, F., Cohen, A., Zhang, P. et al., (2009). Autologous and allogeneic marrow

stromal cells are safe and effective for the treatment of acute kidney

injury. Stem Cells Dev.,18: 475-485.

Togel, F., Hu, Z., Weiss, K., et al. (2005). Administered mesenchymal stem cells

protect against ischemic acute renal failure through differentiation-

independent mechanisms. Am J Physiol Renal Physiol., 289: F31-F42.

Togel, F., Weiss, K., Yang, Y. et al., (2007). Vasculotropic, paracrine actions of

infused mesenchymal stem cells are important to the recovery from acute

kidney injury. Am J Physiol Renal Physiol., 292: F1626-F1635.

Tomonaga, H. (1993). Detection of antibody specific to rifampicin metabolite by

ELISA-mechanism of sensitization by rifampicin. Arerugi., 42(7): 854-863.

Trapnell, C., Pacher, I. and Salzberg, S.I. (2009): “TopHat: discovering splice

junctions with RNA-Seq,” Bioinformatics, 25(9):1105–111.

© COPYRIG

HT UPM

214

Trapnell, C., Williams, B. A., Pertea, G. et al., (2010): “Transcript assembly and

quantification by RNA-Seq reveals unannotated transcripts and isoform

switching during cell differentiation,” Nature Biotechnology, 28(5):511–515.

Tsenova, L., Ellison, E., Harbacheuski, R., Moreira, A.L., Kurepina, N. et al. (2005).

Virulence of selected Mycobacterium tuberculosis clinical isolates in the

rabbit model of meningitis is dependent on phenolic glycolipid produced by

the bacilli. J Infect Dis., 192: 98-106.

Tormin, Ariane., Li, O., Brune, C.J., Walsh, S., Schutz, B., Ehinger, M., Ditzel, N.,

Kassem, M. and Scheding, S. (2011). CD146 expression on primary

nonhematopoietic bone marrow stem cells is correlated with in situ

localization. Blood, 117:5067-5077.

Tostmann, A., Boeree, M.J., Aarnoutse, R.E., de Lange, W.C., van der Ven, A.J. and

Dekhuijzen, R. (2008). Antituberculosis drug-induced hepatotoxicity:

Concise up-to-date review. J. Gastroenterol. Hepatol., 23: 192–202.

Turktas, H., Unsal, M., Tulek, N. and Oruc, O. (1994). Hepatotoxicity of

antituberculosis therapy (rifampicin, isoniazid and pyrazinamide) or viral

hepatitis. Tuber Lung Dis., 75: 58-60.

U.S. Public Health Service. (1969). Hepatic toxicity of pyrazinamide used with

isoniazid in tuberculous patients. Am Rev Respir Dis., 59:13.

Uberti-Foppa, C., De Bona, A., Morsica, G., Galli, L., Gallotta, G., Boeri, E. and

Lazzarin, A. (2003). Pretreatment of chronic active hepatitis C in patients

coinfected with HIV and hepatitis C virus reduces the hepatotoxicity

associated with subsequent antiretroviral therapy. J Acquir Immune Defic

Syndr., 33: 146-152.

Upadhyay, G., Kumar, A. and Singh, M.P. (2007). Effect of Silymarin on pyrogallol-

and rifampicin-induced hepatotoxicity in mouse. Eur. J. Pharmacol., 565:

190-201.

Van Assendelft, A.H. (1986). Renal failure and hemolysis caused by rifampicin.

Tubercle., 67(3): 234-235.

Van, Poll D., Parekkadan, B., Cho, CH., Berthiaume, F., Nahmias, Y., Tilles, A.W.

and Yarmush, M.L. (2008). Mesenchymal stem cell-derived molecules

directly modulate hepatocellular death and regeneration in vitro and in vivo.

Hepatology. 47: 1634-1643.

Venkataramana, N. K., Pal, R., Rao, S. A. et al., (2012). Bilateral transplantation of

allogenic adult human bone marrow-derived mesenchymal stem cells into the

subventricular zone of Parkinson’s disease: a pilot clinical study. Stem Cells

International, 20(12): 12 Article ID 931902.

© COPYRIG

HT UPM

215

Villarino, M.E., Ridzon, R., Weismuller, P.C., Elcock, M., Maxwell, R.M., Meador,

J., Smith, P.J., Carson, M.L. and Geiter, L.J. (1997): Rifampin preventive

therapy for tuberculosis infection: experience with 157 adolescents. Am J

Respir Crit Care Med., 155: 1735-1738.

Viswanatha Swamy, A. H. M., Rucha V. Kulkarni, Koti, B. C., Gadad, P.

C. Thippeswamy, A. H. M. and Aparna Gore (2012). Hepatoprotective Effect

of Cissus quadrangularis Stem Extract Against Rifampicin-induced

Hepatotoxicity in Rats. Indian J Pharm Sci. 74(2): 183–187.

Von Bonin, M., Stolzel, F., Goedecke, A., Richter, K., Wuschek, N., Holig, K.,

Platzbecker, U., Illmer, T., Schaich, M. and Schetelig, J. (2009). Treatment

of refractory acute GVHD with third-party MSC expanded in platelet lysate-

containing medium. Bone Marrow Transplant. 43: 245-251.

WHO (2016): Multidrug-resistant tuberculosis (MDR-TB) 2016 update.

www.who.int/tb/areas-of-work/drugresistant-tb/treatment/resources/en/.

WHO (2016): Global TB report. Pp 1. ISBN9789241565394 (http://www.who.int)

Wang, S. P., Wang, Z. H., Peng, D. Y., Li, S. M., Wang, H. and Wang, X. H. (2012).

Therapeutic effect of mesenchymal stem cells in rats with intracerebral

hemorrhage: reduced apoptosis and enhanced neuroprotection. Molecular

Medicine Reports, 6(4): 848-854.

Warner, S.D. (1975). Nephrotic syndrome in rhesus monkeys. Abstracts of cases

presented at the 25th Annual meeting American college of veterinary

pathologists. Vet. Pathol., 12: 61-73.

Wayne, L.G and Sohaskey, C.D. (2001). Nonreplicating persistence of

Mycobacterium tuberculosis. Annu Rev Microbiol., 55:139e163.

Wessels, C.C. (1941). Tuberculosis in the rat. II. The fate of tubercle bacilli in the

various organs of the rat. Am. Rev Tuberc., 43: 459-474.

Wieser, M., Stadler, G., Jennings, P., Streubel, B., Pfaller, W., Ambros, P., et al.

(2008). hTERT alone immortalizes epithelial cells of renal proximal tubules

without changing their functional characteristics. Am. J. Physiol. Renal

Physiol. 295, F1365–1375.

Williams, G.M. and Iatropoulos, M.J. (2002). Alteration of liver cell function and

proliferation: differentiation between adaptation and toxicity. Toxicol

Pathol., 30: 41-53.

Woolsey, S.J., Beaton, M.D., Choi, Y.H., Dresser, G.K., Gryn, S.E., Kim, R.B. and

Tirona, R.G. (2016). Relationships between endogenous plasma biomarkers

of constitutive cytochrome P450 3A activity and single-time-point oral

midazolam microdose phenotype in healthy subjects. Basic Clin. Pharmacol.

Toxicol., 118: 284–291.

© COPYRIG

HT UPM

216

Wu, X., Xu, H., Zhang, Z., Chang, Q., Liao, S., Linqiang, Z., Li, Y., Wu, D. and

Liang, B. (2016): Transcriptome Profiles Using Next-Generation Sequencing

Reveal Liver Changes in the Early Stage of Diabetes in Tree Shrew (Tupaia

belangeri chinensis). Journal of Diabetes Research, 2016: 1-15, Article ID

6238526.

www.eurostemcell.org lastupdated 12/08/2013, retrieved 14/11/2015. Kidney

disease: how could stem cells help?

Yachi, R., Iqarashi, O. and Kiyose, C. (2010): Protective Effects of Vitamin E

Analogs against Carbon Tetrachloride-Induced Fatty Liver in Rats. J Clinic

Biochem Nutri. 7(2):148-54.

Yagi, H., Soto-Gutierrez, A., Parekkadan, B., Kitagawa, Y., Tompkins, R.G.,

Kobayashi, N. and Yarmush, M.L. (2010). Mesenchymal stem cells:

Mechanisms of immunomodulation and homing. Cell Transplant. 19: 667-

679.

Yan, J. and Xie, W. (2016). A brief history of the discovery of PXR and CAR as

xenobiotic receptors. Acta Pharm. Sin. B, 6: 450–452.

Yan, X., Liu, Y., Han, Q., Jia, M., Liao, L., Qi, M. and Zhao, R.C. (2007). Injured

microenvironment directly guides the differentiation of engrafted Flk-1(+)

mesenchymal stem cell in lung. Exp Hematol., 35: 1466-1475.

Yanardag, H., Caner, M., Gunes, Y. and Uygun, S. (2005). Acute hemolysis and

oligoanuric acute renal failure caused by interrupted. Internet J. Nephrol.,

2(1).

Yang, Y., Wu, F., Ward, T., Yan, F., Wu, Q. et al. (2008). Phosphorylation of

HsMis13 by Aurora B kinase is essential for assembly of functional

kinetochore. The Journal of biological chemistry 283: 26726–26736.

Yanqing Gong, Xuesong Li1, Jie Jin, Liqun Zhou, Yinglu Guo 2016: AB015. The

expression and function of coiled-coil domaincontaining protein 34 in human

bladder carcinoma. Transl Androl. Urol.5:1

Young, D.B., Gideon, H.P. and Wilkinson, R.J. (2009). Eliminating latent

tuberculosis. Trends Microbiol., 17: 183-188.

Younossian, A.B., Rochat, T., Ketterer, J.P., Wacker, J. and Janssens, J.P. (2005).

High hepatotoxicity of pyrazinamide and ethambutol for treatment of latent

tuberculosis. Eur Respir J., 26: 462-464.

© COPYRIG

HT UPM

217

Yu, Y., James, C. F., Chen, Z., Chao, G., Meiwen, J., Tao, Q., Desmond, I. B., Lee,

L., Wenjun, B., Tingting, D., Heng, L., Zhenqiang, S., Wendell, D. J., Carrie,

L. M., William, S. B., Feng, Q., Baitang, N., Yan, L., Huixiao, H., Lei, G.,

Nan, M., Tieliu, S., Kevin, Y. W, Russell, D. W., Yuri N., Stephen, J. W.,

Penelope, D., Christopher, E. M., Weida, T., Jean, T., Danielle, T., Leming,

S. and Charles, W. (2014). A rat RNA-Seq transcriptomic BodyMap across

11 organs and 4 developmental stages. Nature communications, 5(3230):1-

11.

Yu, A.S. and Keeffe, E.B. (2002): Nonalcoholic fatty liver disease. Rev

Gastroenterol Disord., 2: 11-19.

You-Lin, T., Yu-ju, L., Jiunn-Ming, S., Hong-Ren, Y., Mao-Meng, T., Chih-Cheng,

C., Ching-Chou, T., Li-Tung, H. and Chien-Ning, H. (2017). High fat diets

sex-specifically affect the renal transcriptome and program obesity, kidney

injury, and hypertension in the offsprings. Nutrients, 9(357):1-19.

Zager, R.A. (1997). Pathogenetic mechanisms in nephrotoxic acute renal failure.

Semin Nephrol., 17(1): 3-14.

Zajicek, G., Oren, R. and Weinreb Jr, M. (1985). The streaming liver. Liver. 5: 293-

300.

Zappia, A. E., Benvenuto, F., Frassoni, F. and Mancardi, G. (2006). Stem cells in

inflammatory demyelinating disorders: a dual role for immunosuppression

and neuroprotection. Expert Opinion on Biological Therapy, 6(1): 17-22.

Zhang, Q., Sun, X., Ding, J., He, P., Liu, Y., Cheng, H., Zhou, C. and Meng, X.

(2015): Autoserum: An optimal supplement for bone marrow mesenchymal

stem cells of liver-injured rats. Stem Cells International, 1-10.

http://dx.doi.org/10.1155/2015/459580.

Zhang, B., Liu, R., Shi, D., Liu, X., Chen, Y., Dou, X., Zhu, X., Lu, C., Liang, W.

and Liao, L. (2009). Mesenchymal stem cells induce mature dendritic cells

into a novel Jagged-2-dependent regulatory dendritic cell population. Blood.

113: 46-57.

Zhang, W., Ge, W., Li, C., You, S., Liao, L., Han, Q., Deng, W. and Zhao, R.C.

(2004). Effects of mesenchymal stem cells on differentiation, maturation, and

function of human monocyte-derived dendritic cells. Stem Cells Dev., 13:

263-271.

Zhang, W.V., Ramzan, I. and Murray, M. (2007). Impaired microsomal oxidation of

the atypical antipsychotic agent clozapine in hepatic steatosis. J. Pharmacol.

Exp. Ther., 322: 770–777.

Zhao, D. C., Lei, J. X., Chen, R. et al., (2005). Bone marrow-derived mesenchymal

stem cells protect against experimental liver fibrosis in rats. World Journal of

Gastroenterology, 11(22): 3431-3440.

© COPYRIG

HT UPM

218

Zhao, W., Li, J. J., D. Cao, Y. et al., (2012). Intravenous injection of mesenchymal

stem cells is effective in treating liver fibrosis. World Journal of

Gastroenterology, 18(10): 1048-1058.

Zhou, H. P., Yi, D. H., Yu, S. Q. et al., (2006). Administration of donorderived

mesenchymal stem cells can prolong the survival of rat cardiac allograft.

Transplantation Proceedings, 38: (9): 3046-3051.

Zhu, X.Y., Urbieta-Caceres, V., Krier, J.D., Textor, S.C., Lerman, A. and Lerman,

L.O. (2013). Mesenchymal stem cells and endothelial progenitor cells

decrease renal injury in experimental swine renal artery stenosis through

different mechanisms. Stem Cells, 31: 117-125.

Zhu, H., Guo, Z., Jiang, X., Li, H., Wang, X., Yao, H., Zhang, Y. and Mao, N.

(2010). A protocol for isolation and culture of mesenchymal stem cells from

mouse compact bone. Natures protocols, 5(3):550-560.