134
UNIVERSITATIS OULUENSIS MEDICA ACTA D D 1271 ACTA Tuomas Mäkelä OULU 2014 D 1271 Tuomas Mäkelä SYSTEMIC TRANSPLANTATION OF BONE MARROW STROMAL CELLS AN EXPERIMENTAL ANIMAL STUDY OF BIODISTRIBUTION AND TISSUE TARGETING UNIVERSITY OF OULU GRADUATE SCHOOL; UNIVERSITY OF OULU, FACULTY OF MEDICINE, INSTITUTE OF CLINICAL MEDICINE, DEPARTMENT OF SURGERY, DEPARTMENT OF ANAESTHESIOLOGY; INSTITUTE OF BIOMEDICINE, DEPARTMENT OF ANATOMY AND CELL BIOLOGY; INSTITUTE OF DIAGNOSTICS, DEPARTMENT OF DIAGNOSTIC RADIOLOGY; CLINICAL RESEARCH CENTER

OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

ABCDEFG

UNIVERSITY OF OULU P .O. B 00 F I -90014 UNIVERSITY OF OULU FINLAND

A C T A U N I V E R S I T A T I S O U L U E N S I S

S E R I E S E D I T O R S

SCIENTIAE RERUM NATURALIUM

HUMANIORA

TECHNICA

MEDICA

SCIENTIAE RERUM SOCIALIUM

SCRIPTA ACADEMICA

OECONOMICA

EDITOR IN CHIEF

PUBLICATIONS EDITOR

Professor Esa Hohtola

University Lecturer Santeri Palviainen

Postdoctoral research fellow Sanna Taskila

Professor Olli Vuolteenaho

University Lecturer Veli-Matti Ulvinen

Director Sinikka Eskelinen

Professor Jari Juga

Professor Olli Vuolteenaho

Publications Editor Kirsti Nurkkala

ISBN 978-952-62-0653-0 (Paperback)ISBN 978-952-62-0654-7 (PDF)ISSN 0355-3221 (Print)ISSN 1796-2234 (Online)

U N I V E R S I TAT I S O U L U E N S I S

MEDICA

ACTAD

D 1271

ACTA

Tuomas M

äkelä

OULU 2014

D 1271

Tuomas Mäkelä

SYSTEMIC TRANSPLANTATION OF BONE MARROW STROMAL CELLSAN EXPERIMENTAL ANIMAL STUDY OF BIODISTRIBUTION AND TISSUE TARGETING

UNIVERSITY OF OULU GRADUATE SCHOOL;UNIVERSITY OF OULU,FACULTY OF MEDICINE,INSTITUTE OF CLINICAL MEDICINE,DEPARTMENT OF SURGERY,DEPARTMENT OF ANAESTHESIOLOGY;INSTITUTE OF BIOMEDICINE,DEPARTMENT OF ANATOMY AND CELL BIOLOGY;INSTITUTE OF DIAGNOSTICS,DEPARTMENT OF DIAGNOSTIC RADIOLOGY;CLINICAL RESEARCH CENTER

Page 2: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the
Page 3: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

A C T A U N I V E R S I T A T I S O U L U E N S I SD M e d i c a 1 2 7 1

TUOMAS MÄKELÄ

SYSTEMIC TRANSPLANTATION OF BONE MARROW STROMAL CELLSAn experimental animal study of biodistribution and tissue targeting

Academic dissertation to be presented with the assent ofthe Doctoral Training Committee of Health andBiosciences of the University of Oulu for public defencein Auditorium 1 of Oulu University Hospital, on 19December 2014, at 12 noon

UNIVERSITY OF OULU, OULU 2014

Page 4: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

Copyright © 2014Acta Univ. Oul. D 1271, 2014

Supervised byProfessor Petri LehenkariProfessor Tatu Juvonen

Reviewed byProfessor Yrjö KonttinenDocent Susanna Miettinen

ISBN 978-952-62-0653-0 (Paperback)ISBN 978-952-62-0654-7 (PDF)

ISSN 0355-3221 (Printed)ISSN 1796-2234 (Online)

Cover DesignRaimo Ahonen

JUVENES PRINTTAMPERE 2014

OpponentProfessor Ari Harjula

Page 5: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

Mäkelä, Tuomas, Systemic transplantation of bone marrow stromal cells. Anexperimental animal study of biodistribution and tissue targetingUniversity of Oulu Graduate School; University of Oulu, Faculty of Medicine, Institute ofClinical Medicine, Department of Surgery, Department of Anaesthesiology; Institute ofBiomedicine, Department of Anatomy and Cell Biology; Institute of Diagnostics, Department ofDiagnostic Radiology; Clinical Research CenterActa Univ. Oul. D 1271, 2014University of Oulu, P.O. Box 8000, FI-90014 University of Oulu, Finland

Abstract

Bone marrow mesenchymal stromal cells (MSCs) and mononuclear cells (BM-MNCs) haveshown great therapeutic potential in various clinical settings. Although intravasculartransplantation of the cells constitutes the optimal delivery route, massive pulmonary entrapment,with the threat of embolization, remains a major obstacle for using this type of therapy. Becausepulmonary entrapment is at least partially mediated by adhesion molecules, cell surfacemodification could enhance pulmonary passage.

We used a porcine model of allogeneic MSC and autologous BM-MNC transplantation andradionuclide labelling to track the cells. The role of the transplantation route on lung entrapment,biodistribution, safety and BM-MNC targeting to the injured brain was studied. Effects of pronasedetachment on the lung passage of MSCs were studied in porcine and murine models; a rat modelof acute limb injury was used to further evaluate tissue targeting. Treatment with pronase to detachcell surface molecules and the effect on stem cell potential was assessed in vitro.

Intra-arterial administration of MSCs diminishes their lung deposition; intravasculartransplantation did not cause pulmonary embolisms. Intra-arterially transplanted BM-MNCs didnot reach the brain in significant numbers. Transient proteolytic modification of MSCs withpronase decreased lung accumulation and tissue targeting without affecting their therapeuticcharacteristics.

Intra-arterial transplantation increases lung passage of MSCs. Although thromboembolicevents were not observed, further studies are warranted to ensure the safety of this route of MSCdelivery. Pronase detachment is a promising method to enhance the potential of systemic MSCtherapies.

Keywords: biodistribution, imaging, lung entrapment, stem cells

Page 6: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the
Page 7: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

Mäkelä, Tuomas, Luuytimen mesenkymaalisten kantasolujen systeeminenannostelu. Koe-eläintyö kudosjakautumisesta ja -kohdentumisestaOulun yliopiston tutkijakoulu; Oulun yliopisto, Lääketieteellinen tiedekunta, Kliinisenlääketieteen laitos, Kirurgia, Anestesiologia; Biolääketieteen laitos, Anatomia ja solubiologia;Diagnostiikan laitos, Radiologia; Kliinisen tutkimuksen keskusActa Univ. Oul. D 1271, 2014Oulun yliopisto, PL 8000, 90014 Oulun yliopisto

Tiivistelmä

Luutytimen mesenkymaaliset kantasolut (MSC) ja mononukleaariset solut (BM-MNC) ovatosoittautuneet tehokkaiksi useissa kliinisissä käyttöaiheissa. Solujen systeeminen annosteluverenkiertoon olisi käytännön kannalta paras soluterapian toteutukseen, mutta solujen merkittä-vä taipumus jäädä keuhkoihin loukkuun ja veritulppariski muodostavat haasteen. Keuhkohakeu-tumisen tiedetään ainakin osin johtuvan solujen pintamolekyyleistä ja näiden muokkaaminenvoisi parantaa solujen keuhkoläpäisevyyttä.

Tutkimuksessa käytettiin koe-eläimenä sikaa, jolle istutettiin systeemisesti allogeenisiamesenkymaalisia kantasoluja tai autologisia luuytimen mononukleaarisia soluja; solujen kudos-hakeutumisen seuranta toteutettiin isotooppileimauksella- ja kuvannuksella. Tutkimuksessa arvi-oitiin annostelureitin vaikutusta keuhkoläpäisevyyteen, solujen kudojakautumista, toimenpiteenturvallisuutta sekä mononukleaarisolujen hakeutumista vaurioituneeseen aivokudokseen. Pro-naasikäsittelyn vaikutusta mesenkymaalisten kantasolujen keuhkoläpäisevyyteen arvioitiin sika-ja hiirimallissa; rotan raajavauriomallia käytettiin lisäksi pronaasin kudoshakeutumisvaikutus-ten arvioimiseen. Pronaasikäsittelyn vaikutuksia solujen pintarakenteisiin ja toiminnallisuuteenarvioitiin in vitro- kokeissa.

Mesenkyymalisten kantasolujen annostelu valtimonsisäisesti paransi solujen keuhkoläpäise-vyyttä; tutkimuksissa käytetyt solut eivät aiheuttaneet keuhkoveritulppia. Valtimonsisäisestiannostellut mononukleaarisolut eivät hakeutuneet vaurioituneeseen aivokudokseen sikamallissa.Pronaasikäsittely muovasi solujen pintaproteiineja palautuvasti ja tämä lisäsi huomattavastimesenkymaalisten kantasolujen keuhkoläpäisevyyttä ja kudoshakeutumista vaikuttamatta solu-jen toiminnallisuuteen.

Mesenkymaalisten kantasolujen annostelu valtimonsisäisesti voi parantaa solujen keuhkolä-päisevyyttä. Tutkimuksessa ei todettu keuhkoveritulppaa tai muita tromboembolisia tapahtumia,mutta lisätutkimuksia tarvitaan MSC- terapian turvallisuuden takaamiseksi. Pronaasikäsittelyntulokset mesenkymaalisten kantasolujen systeemisen annostelun parantamisessa olivat lupaavia.

Asiasanat: kantasolut, keuhkoläpäisevyys, kudosjakautuminen, kuvantaminen

Page 8: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the
Page 9: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

to Kristiina

Page 10: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

8

Page 11: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

9

Acknowledgements

The present study has been conducted at the Cardiothoracic Research Laboratory

of the Department of Surgery, Oulu University Hospital, during the years 2007-

2014.

I am most grateful to my professors, Petri Lehenkari and Tatu Juvonen, for

their continued support during this project. Closely collaborating with Professor

Lehenkari has been a true pleasure. I consider Professor Lehenkari to be an

excellent and innovative scientist; however, the most striking feature about him is

his ability to create hope where none exists. I am most thankful to Professor

Juvonen for recruiting me to his renowned group of experimental surgeons. I have

had the privilege to benefit from his support and his long-time experience with

experimental models, as well as the technical knowledge of his well-established

research group.

I am also most grateful to Professor Yrjö Konttinen and Docent Susanna

Miettinen for acting as the pre-examiners of this manuscript. Finding experts to

review such a specific research subject is not easy and I was fortunate enough that

they agreed to take on this project. Their comments greatly improved this work. I

would also like to thank the members of my follow-up group for their valuable

contribution, Docent Jari Satta and Docent Pekka Rainio.

This project required the effort, sacrificing of free time, patience, brainpower and

skill of many colleagues. I consider that working long hours using such a

demanding experimental model is the ultimate form of teamwork. I would like to

thank Fredrik Yannopoulos, Kirsi Heikkinen, Oiva Arvola, Henri Haapanen and

Johanna Herajärvi for their contributions to this project and the sacrifices that

they made. With the right people, even the most challenging tasks can be

accomplished with ease (we even managed to have a laugh every now and then).

I am also grateful to the older colleagues of our experimental group, Jussi

Mäkelä, Hanna Jensen, Eija Rimpiläinen and Sanna Meriläinen, with whom I had

the privilege to work on their projects. They taught me the principles behind our

experimental models and adopted me as one of their own when I first arrived to

the lab. I also wish to thank our group’s senior reseachers for their valuable

assistance on the project. Docent Vesa Anttila, docent Kai Kiviluoma and

Professor Roberto Blanco all offered expertise in their own clinical fields, which

greatly benefited this project.

I would especially like to thank Reijo Takalo for his collaborative efforts and

expertise in radionuclide medicine. Creating our experimental model and

Page 12: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

10

interpreting the data was demanding, but noteworthy. Aira Karjalainen and Leila

Mäkelä, both staff members of the Radionuclide Department, offered their

precious assistance. The rest of the radionuclide department also deserves

recognition for tolerating the demanding arrangements for the experiments that

Aira and Leila had to conduct in addition to their daily routines. I am grateful to

the Department of Radiology and Lauri Ahvenjärvi in particular, for arranging the

safety control imaging studies.

Our collaborators in the Finnish Red Cross Blood Service, Docent Erja

Kerkelä and Johanna Nystedt, along with their local team, deserve my deepest

gratitude. Their in-depth knowledge of the bone marrow therapeutic field elevated

our study design and the quality of our written products to a whole new level. It

was an honour for me to collaborate with their team on the publication of my

pronase modification studies.

The entire team of our stem cell laboratory deserves my deepest thanks for

the work involving cell cultivation and processing. Docent Siri Lehtonen, Minna

Savilampi, Henna Ek and Elisa Lehtilahti, your adaptability to our demanding

schedules was amazing and you were always able to provide high-quality cell

products for use in our experiments. Pasi Lepola deserves thanks for carrying out

the electrophysiologic analysis with such a professional touch, despite the long

working hours. Pasi Ohtonen and Risto Bloigu are thanked for their expert

statistical consultation.

A very special thanks goes to Seija Seljänperä for acting as the mother of our

small research family. Docent Hanna-Marja Voipio, Sakari Laaksonen, Veikko

Lähteenmäki and the other members of the Animal Research Centre staff are

warmly acknowledged for providing use of their fine facility, along with their

skilled animal care.

I am grateful to all my friends for their precious company during all these

years and for offering a chance to relax and participate in athletic activities

together. My thanks also go to my current working colleagues in the Lappish

Central Hospital’s Department of Surgery for providing a supportive atmosphere

while I was completing this project and my surgical residency simultaneously.

Pirkko, Mikko, Matti, Kirsti, Sonja, Konsta and Milja, my beloved parents

and family, and not to forget my dear in-laws Marjatta, Heikki and Tommi, thank

you for your endless support. To have all of you in my life is bliss.

Finally, my deepest and sincerest thanks goes to the one person that has

sacrificed even more for this project than I, Kristiina, my love. Countless times

we have had to plan our life according to the demands of this project, with

Page 13: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

11

countless hours having to be spent apart. Still, you never lost hope and always

provided constant support. Even more amazingly, you still agreed to be my wife.

To have you by my side makes me the happiest man alive. You are the one who

has made all this possible.

This work was financially supported by the Finnish Red Cross Blood Service

and the Finnish Foundation for Cardiovascular Research.

Oulu, October 2014 Tuomas Mäkelä

Page 14: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

12

Page 15: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

13

Abbreviations

APC Antigen presenting cells

BBB Blood brain barrier

BCAS Bilateral common carotid artery stenosis

BDNF Brain-derived neurotrophic factor

BNGF β-nerve growth factor

BM-MNC Bone marrow mononuclear cell

CBF Cerebral blood flow

CLSM Confocal laser scanning microscopy

CPB Cardio-pulmonary bypass

CSPG-4 Chondroitin sulfate proteoglycan-4

CT Computed tomography

DC Dendritic cells

ECG Electrocardiography

EEG Electroencephalography

ELISA Enzyme-linked immunosorbent assay

EPC Endothelial progenitor cells

ESC Embryonic stem cells

FACS Fluorescent-activated cell sorting

FN Fibronectin

GM-CSF Granulocyte-macrophage colony-stimulating factor

Gmean Geometric mean

GvHD Graft versus host disease

HCA Hypothermic circulatory arrest

HGF Hepatocyte growth factor

hMSC Human marrow stromal cell (mesenchymal stem cell)

mMSC Mouse marrow stromal cell

HSC Hematopoietic stem cell

hUC-MSC Human umbilical cord-derived marrow stromal cell

IA Intra-arterial

IBMIR Instant blood-mediated inflammatory reaction

IDO Indoleamine 2,3-dioxygenase

IV Intravenous

MCA Middle cerebral artery

MCAO Middle cerebral artery occlusion

mDC Myeloid dendritic cells

Page 16: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

14

MI Myocardial infarction

MRI Magnetic resonance imaging

NGF Nerve growth factor

NK Natural killer cells

NPC Neural progenitor cells

NSC Neural stem cells

MSC(s) Marrow stromal cell (mesenchymal stem cell)

OI Osteogenesis imperfecta

OPC Oligodendrocyte progenitor cells

pDC Plasmacytoid dendritic cells

PELOD Pediatric logistic organ dysfunction score

PET Positron emission tomography

PID Post-ischemic day

ROI Region of interest

SDF-1 Stromal cell-derived factor-1α

SPECT Single photon emission tomography

SPECT/CT Single photon emission tomography- computed tomography

combination

Tc99m-HMPAO Technetium99m-hydroxymethylpropylene amine oxime

TBI Traumatic brain injury

TF Tissue factor

TUNEL Terminal deoxynucleotidyltransferase-mediated dUTP-biotin

nick-end labelling

VOI Volume of interest

Page 17: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

15

List of original publications

This thesis is based on the following articles, which are referred to in the text by

their Roman numerals:

I Mäkelä T, Takalo R, Arvola O, Haapanen H, Yannopoulos F, Blanco R, Ahvenjärvi L, Lehtonen S, Kiviluoma K, Kerkelä E, Nystedt J, Juvonen T & Lehenkari P. (2014) Safety and Biodistribution Study of Bone Marrow Derived Mesenchymal Stromal Cells and Mononuclear Cells and the Impact of the Administration Route in an Intact Porcine Model. Manuscript

II Mäkelä T, Yannopoulos F, Alestalo K, Mäkelä J, Lepola P, Anttila V, Lehtonen S, Kiviluoma K, Takalo R, Juvonen T & Lehenkari P. (2013) Intra-Arterial Bone Marrow Mononuclear Cell Distribution after Experimental Global Brain Ischemia Scand Cardiovasc J. 47(2):114-20.

III Kerkelä E, Hakkarainen T, Mäkelä T, Kilpinen L, Lehtonen S, Ritamo I, Pernu R, Pietilä M, Takalo R, Nikkilä J, Tikkanen J, Juvonen T, Laitinen S, Valmu L, Lehenkari P & Nystedt J. (2013) Transient Proteolytic Modification of Mesenchymal Stromal Cells Decreases Lung Entrapment in vivo. Stem Cells Translational Medicine. 2(7):510-20.

Page 18: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

16

Page 19: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

17

Table of Contents

Abstract

Tiivistelmä

Acknowledgements 9 Abbreviations 13 List of original publications 15 Table of Contents 17 1 Introduction 21 2 Review of the literature 23

2.1 Stem cells ................................................................................................ 23 2.1.1 Bone marrow stromal cells ........................................................... 23 2.1.2 Bone marrow mononuclear cells .................................................. 24

2.2 Mechanisms behind MSC therapeutic properties .................................... 25 2.2.1 Paracrine effects of MSCs ............................................................ 26 2.2.2 Immunomodulation ...................................................................... 28 2.2.3 Effects on T-cells .......................................................................... 30

2.3 Clinical applications of MSCs ................................................................ 32 2.3.1 Ischemic brain injury and stroke .................................................. 33 2.3.2 Other indications .......................................................................... 40

2.4 Intravascular transplantation of bone marrow-derived cells ................... 42 2.4.1 The role of direct transplantation .................................................. 43 2.4.2 Passive biodistribution after intravascular transplantation ........... 44 2.4.3 Active biodistribution by chemotaxis and migration .................... 44 2.4.4 Biodistribution tracking methods ................................................. 45 2.4.5 Acute phase biodistribution .......................................................... 49 2.4.6 Long-term biodistribution ............................................................ 50

2.5 Challenges of intravascular MSC transplantation ................................... 50 2.5.1 Periprocedural damage to MSCs .................................................. 51 2.5.2 Pulmonary adhesion and the lung barrier ..................................... 54 2.5.3 Thromboembolic events ............................................................... 55 2.5.4 Immunologic reactions ................................................................. 58 2.5.5 Development of malignancies ...................................................... 59 2.5.6 Ectopic tissue formation ............................................................... 59

2.6 Optimisation of intravascular MSC transplantation ................................ 60 2.6.1 Type of injury ............................................................................... 60 2.6.2 Cell type ....................................................................................... 61

Page 20: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

18

2.6.3 Systemic transplantation route ...................................................... 62 2.6.4 Timing of transplantation ............................................................. 62 2.6.5 Cell dose ....................................................................................... 63 2.6.6 Pronase detachment ...................................................................... 65 2.6.7 Vasodilatators ............................................................................... 66 2.6.8 Anticoagulative agents ................................................................. 66 2.6.9 Cellular engineering and culture conditions ................................. 67

3 Aims of the study 69 4 Materials and methods 71

4.1 Experimental animals (Studies I, II, III).................................................. 71 4.2 Perioperative protocol of porcine model (Studies I, II and III) ............... 71 4.3 Porcine MSCs and BM-MNCs (Studies I, II and III) .............................. 72 4.4 Human MSCs (Study III) ........................................................................ 72 4.5 Rat MSCs (Study III) .............................................................................. 73 4.6 Pronase detachment and cell viability (Study III) ................................... 73 4.7 Cell surface analysis by mass spectrometry (Study III) .......................... 73 4.8 Cell surface analysis by flow cytometry (Study III) ............................... 74 4.9 T-cell proliferation assay (Study III) ....................................................... 74 4.10 Angiogenesis assay (Study III) ............................................................... 75 4.11 In vitro rolling assay (Study III) .............................................................. 75 4.12 Labelling of the porcine cells (Studies I, II and III) ................................ 75 4.13 Intravenous transplantation (Studies I, II and III) ................................... 76 4.14 Intraventricular transplantation (Study I) ................................................ 76 4.15 Ischemic brain insult (Study II) ............................................................... 77 4.16 Radionuclide imaging (Studies I, II and III) ........................................... 77 4.17 Image and data analysis (Studies I, II and III) ........................................ 77 4.18 Pulmonary embolism CT- scan (Study I) ................................................ 78 4.19 Tissue biopsies (Studies I, II and III) ...................................................... 78 4.20 Electroencephalography (Study II) ......................................................... 79 4.21 Biodistribution of human MSCs in a mouse model (Study III) .............. 79 4.22 Biodistribution of allogeneic BM-MSCs in a rat model of

inflammation (Study III) ......................................................................... 80 4.23 Statistical analysis (Study I, II, and III)................................................... 80

5 Results 83 5.1 IA versus IV transplantation of BM-MNCs and MSCs (Study I) ........... 83

5.1.1 Computed tomography ................................................................. 83 5.1.2 Haemodynamic data ..................................................................... 87

Page 21: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

19

5.2 BM-MNC transplantation for global ischemic brain injury during

first 24 hours (Study II) ........................................................................... 88 5.2.1 Proportional counts from biopsies ................................................ 89 5.2.2 Haemodynamic and biochemical data .......................................... 93

5.3 Effects of MSC pronase detachment on lung clearance and organ

targeting (Study III)................................................................................. 94 5.3.1 The effect of pronase detachment on cell-surface protein

epitopes......................................................................................... 94 5.3.2 MSC Functionality ....................................................................... 95 5.3.3 MSCs and lung clearance ............................................................. 96 5.3.4 MSC targeting to an area of injury ............................................... 97

6 Discussion 99 6.1 Biodistribution after intravascular transplantation .................................. 99 6.2 Lung entrapment ................................................................................... 100 6.3 The effect of transplantation route on biodistribution ........................... 101 6.4 Safety assessment .................................................................................. 101 6.5 IA-transplanted BM-MNCs’ lack of homing to ischemic brain ............ 103 6.6 The effect of transplantation timing on homing .................................... 104 6.7 The effect of pronase detachment on MSC lung clearance ................... 105 6.8 Clinical applications .............................................................................. 107 6.9 Limitations of the study ........................................................................ 109 6.10 Summary ................................................................................................110

7 Conclusion 111 References 113 Original publications 129

Page 22: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

20

Page 23: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

21

1 Introduction

Treatment with stem cells is one of the most promising novel forms of therapies

and is slowly gaining acceptance into clinical practice. An increasing number of

studies have documented successful therapeutic applications of mesenchymal

stromal cells (MSCs) and bone marrow mononuclear cells (BM-MNCs) in both

preclinical and clinical settings. Restorative and immunomodulatory properties of

these cells have debuted in the treatment of various socio-economically high-

impact clinical conditions, such as ischemic brain injury.

Previous research has been understandably directed towards proving the

therapeutic effectiveness of these cells in various clinical conditions and

preclinical success stories have been gradually translated into human use.

However, MSCs remain indisputable as an experimental form of treatment,

because their basic characteristics and behaviour after transplantation are still

incompletely understood.

Intravascular transplantation of MSCs has served as the major delivery route

in this field due to its non-invasiveness and applicability in a variety of clinical

conditions. Researchers have faced unexpected problems with seemingly simple

intravenous and intra-arterial administration of cells, such as undesirable patterns

of organ distribution and failure to reach the therapeutic target. Furthermore,

alarming reports of possible adverse effects resulting from embolus formation

have also been reported. Before MSCs are ultimately able to enter routine clinical

practice as a therapeutic tool, these issues have to be thoroughly researched and

resolved.

The theoretical potential of MSCs, as well as other stem cells, is undeniable

and we are approaching a time when these cells will become available as a

standard therapeutic choice. However, we already have knowledge of what MSCs

can do; now it is of the utmost importance to explore how to actually use them.

This study evaluates the issues of tissue targeting and safety associated with

systemic transplantation of MSCs and offers novel solutions to enhance this form

of cellular therapy. MSC therapy has been investigated in multiple therapeutic

indications, all of which have shared, but also indication-specific, challenges.

Ischemic brain injury is a particularly interesting application of MSCs, as major

issues have been observed when applying MSCs for this indication. Ischemic

brain injury was therefore chosen as an example of an experimental target of the

therapeutic cells.

Page 24: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

22

Page 25: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

23

2 Review of the literature

2.1 Stem cells

Stem cells are capable of self-renewal and multi-lineage differentiation (Till &

McCulloch 1961). As a result of these properties, stem cells are able to

differentiate into multiple cellular lines, while renewing themselves. Two main

types of stem cells exist: (1) embryonic stem cells, which are found during the

early stages of development, appearing in the inner mass of blastocysts, and (2)

adult stem cells, which can be isolated from various tissues and have a more

limited differentiation capacity. Also, embryonic fetal cells from the genital ridge

and tissue-specific sites can be considered as separate stem cell categories

(Arvidson et al. 2011).

Plastic potency of stem cells determines the capacity of the stem cell

populations to differentiate into other cell types. Totipotent cells are capable of

differentiating into all embryonic and non-embryonic cell types of the human

body and are formed after fusion of spermatocytes and oocytes (Tarkowski 1959).

Totipotent stem cells are able to create an entire organism. Pluripotent stem cells,

which also appear during early stages of embryonic development, are capable of

differentiating into any cell type like totipotent stem cells, but lack the capacity of

forming an entire organism by themselves. Multipotent stem cells are a more

differentiated type of stem cell having less plastic capacity, giving rise to a

limited number of cells of a certain tissue type, such as blood cells or neural cells.

Totipotent, pluripotent and multipotent stem cells arise at different stages of

embryonic development and generally specialise further as a function of time,

with their plastic capacity gradually diminishing.

Adult stem cells are non-specialised multipotent cells present in specialised

tissue that are capable of renewing themselves and forming types of cells of the

tissue from which they originate (Till & McCulloch 1961). Examples of adult

stem cell types include hematopoietic (HSC) (Till & McCulloch 1961, Becker et al. 1963) and neural stem cells (Stemple & Anderson 1992)

2.1.1 Bone marrow stromal cells

It has been discovered that bone marrow serves as a source of non-haematopoietic

multipotent stem cells, called bone mesenchymal stem cells, or now more

Page 26: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

24

commonly known as marrow stromal cells (MSCs) (Caplan 1991). This

population of cells was originally described by Friedenstein et al. and were first

identified as fibroblast colonies (Friedenstein et al. 1970). These cells are

multipotent stem cells capable of differentiating into osteoblasts, chondrocytes,

adipocytes and myoblasts (Pittenger et al. 1999, Prockop 1997). There is also

evidence that MSCs can even differentiate into astrocytes (Kopen et al. 1999),

glial cells (Sanchez-Ramos et al. 2000) and neurons (Deng et al. 2001, Mezey et al. 2000, Sanchez-Ramos et al. 2000, Woodbury et al. 2000).

There still does not exist a single MSC marker or identifiable characteristic,

which could be used for their unequivocal identification and isolation. Therefore,

there is some controversy as to how to define MSCs, but the commonly accepted

minimum criteria for MSCs include the ability to adhere to plastic and the

potential for in vitro trilineage differentiation into adipogenic, chondrogenic, and

osteogenic cells. In addition, MSCs must also display CD105, CD73 and CD90,

and lack CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR surface

molecules (Dominici et al. 2006).

Although bone marrow is the first described and mostly commonly used

source of MSCs in clinical and preclinical applications, several other tissues serve

as a source of MSCs, including adipose tissue (Zuk et al. 2002), human placenta

(In't Anker et al. 2004), skin (Shih et al. 2005), human umbilical cord blood

(Erices et al. 2000), human breast milk (Patki et al. 2010), dental pulp (Gronthos et al. 2000), amniotic fluid (Nadri & Soleimani 2007), umbilical cord

perivascular cells (Sarugaser et al. 2005), human umbilical cord Wharton’s jelly

(Wang et al. 2004) and the synovial membrane of the knee joint (De Bari et al. 2001). The MSCs acquired from different sources are likely to have somewhat

varying differentiation potential and cell characteristics.

2.1.2 Bone marrow mononuclear cells

The mononuclear cell fraction isolated from bone marrow aspirates is commonly

referred to as bone marrow mononuclear cells (BM-MNCs). The mononuclear

fraction is a heterogenous mixture containing several populations of stem cells,

such as hematopoietic stem cells, endothelial progenitor cells, MSCs and others

with a greater differentiation capacity (Cuende et al. 2012).

Although BM-MNCs are not technically stem cells, as the population contains

mostly cell types other than MSCs, they are utilized in the same manner as

cultivated MSCs. The main reason use of BM-MNCs have become so popular in

Page 27: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

25

this field of study is their proven effectiveness in treating various conditions

(including ischemic brain injury) (Iihoshi et al. 2004, Kamiya et al. 2008, Fujita et al. 2010, de Vasconcelos Dos Santos et al. 2010), their easy availability

through a relatively non-invasive puncture procedure and the possibility of

isolating this fraction in just a few hours, which allows cellular interventions in

acute conditions without the need of expansion in cell cultures. The possibility of

rapid isolation and re-transplantation makes BM-MNCs a competitive choice in

various clinical conditions, where better therapeutic results could be expected by

avoiding delays (Chen et al. 2001b, Barbash et al. 2003, Iihoshi et al. 2004, de

Vasconcelos Dos Santos et al. 2010, Yang et al. 2011). In addition, isolation and

re-transplantation is inherently autologous, thus evading a number of possible

immunologic, infection, prion and xenogenic culture medium issues compared to

transplantation of cultured allogeneic or xenogeneic donor cells.

This review concentrates on research conducted using MSCs. Although

studies were also conducted using BM-MNCs, the results obtained are not

directly translatable to MSCs due to the different composition of these two

cellular populations. However, if positive results are shown with MSCs, it can be

hypothesised that BM-MNCs could also show similar therapeutic potential in that

particular clinical disorder. Application of BM-MNCs instead of MSCs should be

considered if rapid deployment of the cellular product is required due to the

nature of the event being treated (e.g. acute ischemic brain injury).

2.2 Mechanisms behind MSC therapeutic properties

A common belief with MSC therapy is that they, as cells capable of division and

transdifferentiation, could serve as a replacement for damaged cells, thereby

forming new tissue (Salem & Thiemermann 2012). If MSCs could be shown to

have the ability to differentiate into functional cells of the damaged, or partially

lost and thus cell-depleted target organs (e.g. cardiomyocytes in myocardial

infarction as reported by Orlic et al. 2001), they could theoretically regenerate

new tissue, thereby restoring normal organ function. However, in case of

myocardial infarction, present studies have challenged this theory because

although the transplanted MSCs start to express certain characteristics of native

cardiomyocytes, they do not differentiate into fully mature, functional

cardiomyocytes (Rose et al. 2008).

Using ischemic brain injury as an example of a therapeutic target of MSCs,

recent studies have reported the localization of only very low numbers of cells in

Page 28: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

26

ischemic brain areas during the subacute phase of the developing brain injury

(Harting et al. 2009, Keimpema E. et al. 2009). Poor survival of transplanted

MSCs has been reported (Coyne et al. 2006). Although bone marrow MSCs are

capable of differentiating into astrocytes (Kopen et al. 1999), glia (Bossolasco et al. 2005) and neurons (Mezey et al. 2000, Sanchez-Ramos et al. 2000, Woodbury et al. 2000, Deng et al. 2001, Bossolasco et al. 2005), it appears that the

transplanted MSCs found in the lesional area express neurotrophic factors, but do

not become fully active neurons; however, they may still have therapeutic

potential (Koh et al. 2008). Interestingly, even the transient presence of a small

number of MSCs resulted in a significant reduction in the size of a focal ischemic

brain lesion (Keimpema E. et al. 2009). It thus seems unlikely that the restorative

effect of stem cells is largely based on direct reformation of tissue by the

transdifferentiation of the stem cells at the site of injury.

This theory is supported by a common observation in experimental studies

that the beneficial effects observed after stem cell transplantation are seen far

earlier in ischemic lesions than would be possible by tissue regeneration mediated

by any cell (reviewed by Chopp & Li 2002). Stem cell differentiation and

regeneration may be partially responsible for the therapeutic benefit of MSCs, but

mechanisms other than differentiation and cellular replacement are currently

considered to be of fundamental importance.

2.2.1 Paracrine effects of MSCs

The function of a living organism is based on cellular signalling. Cells constantly

adapt and react to fluctuations in their microenvironments, such as differences in

oxygen content and pH. Cells also react to or receive signals that are generated by

themselves in an intra- or autocrine mode, or by other cells of the organism. Cells

can interact with adjacent cells via direct cell-to-cell contact, which is called

juxtacrine signalling. Cells can also interact with other cells over long distances

by secreting soluble factors. The term paracrine signalling involves local

interactions, while the term endocrine signalling involves systemic interactions.

Secretion of salivary peptide hormones, which affects the gastrointestinal tract, is

an example of an exocrine mode of action. The transmitters of cellular signalling

are mediated by a variety of types of molecules, such as hormones,

neurotransmitters and cytokines. In addition to soluble molecules, cells can also

use direct cell-to-cell and cell-to-extracellular matrix contacts for signalling with

Page 29: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

27

their environment. Furthermore, various physiochemical factors play important

roles in regulating cellular behaviour (Kaivosoja et al. 2012).

Cumulative evidence suggests that the effects of MSC therapy, at least in

part, arise from indirect paracrine mechanisms (Salem & Thiemermann 2012). It

has even been shown that administration of medium conditioned with human

MSCs is able to reduce infarct size in a porcine model (Timmers et al. 2007). It is

likely that the paracrine effects of MSCs arise from multiple functioning secreted

substances rather than a single one. This collection of MSC-derived mediators

has recently been referred to as the MSC secretome (Ranganath et al. 2012).

Angiogenesis

In preclinical studies, BM-MNCs have been shown to increase vascular

endothelial growth factor (VEGF) concentration in global (Fujita et al. 2010) and

focal (Chen et al. 2003) ischemic brain injury. VEGF secretion, which promotes

angiogenesis, has been suggested to be a potential mechanism in bone marrow-

derived cell-induced brain protection. VEGF singularly has been shown to affect

the functional outcome in an animal model of cerebral ischemia and was found to

increase blood-brain-barrier (BBB) permeability (Zhang et al. 2000). MSCs have

also been shown to induce angiogenesis in ischemic myocardium (Martens et al. 2006). VEGF-induced angiogenesis could thus reasonably account for some of

the MSC therapeutic properties. Additionally, the number of Ki-67 positive

proliferating cells increased in a model of ischemic brain injury, implying the

induction of angiogenesis (Shen et al. 2006).

Neuroregenerative effects

MSC administration has been demonstrated to increase synaptophysin expression

and the number of oligodendrocyte progenitor cells in the ischemic border or

twilight zone, resulting in improved recovery, which suggests facilitation of

neuronal remodelling (Shen et al. 2006). MSCs may exhibit anti-apoptotic

properties and can protect neurons against apoptotic stress by stimulation of

endogenous survival signalling in neurons (Isele et al. 2007). Human MSCs have

been demonstrated to secrete brain-derived neurotrophic factor (BDNF) (Chen et al. 2002, Crigler et al. 2006), nerve growth factor (NGF) (Chen et al. 2002,

Crigler et al. 2006) and hepatocyte growth factor (HGF) (Chen et al. 2002). It has

been suggested that the neurorestorative effects of MSCs are also based on other

Page 30: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

28

factors, in addition to BDNF and NGF, as the effects observed in vitro are more

prominent than those which could be explained by the concentrations of these

secreted neurotrophic factors. Indeed, MSCs have been shown to express a large

number of other neuroregulatory factors (Crigler et al. 2006). MSCs may also be

able to stimulate endogenous neural stem cells (Galindo et al. 2011).

Fig. 1. Marrow stromal cell immunomodulatory effects on different immune cell types.

(original figure by Petri Lehenkari).

2.2.2 Immunomodulation

MSCs affect the immunologic system in a number of different ways, most of

which suppress the system. This phenomenon can be exploited in numerous

therapeutic applications in which the MSC therapy targets, at least partially,

pathological conditions mediated by the innate and adaptive immune system.

These conditions occur in various forms of ischemic organ injury, where a major

proportion of the injury is due to reperfusion of the organ after cessation of blood

Page 31: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

29

flow, resulting in a hyper-inflammatory response caused by an influx of white

blood cells (Rao et al. 2014). Graft-versus-host-disease (GvHD) and organ

transplantation rejection are somewhat more obvious targets of MSC therapy,

because the immune system is responsible for the pathogenesis of these

conditions.

Mediators of MSC immunologic effects

It is commonly accepted in the literature that many of the immunomodulatory

effects of MSCs are based on direct cell-to-cell contact or the secretion of soluble

factors. Candidates for MSC mediators are many and it is likely that their

influence is based on a collection of different soluble and cell membrane-bound

substances, rather than a single entity, or in other words, on the MSC secretome

(Ranganath et al. 2012).

Many of the immunomodulatory effects of human MSCs have been

suggested to be mediated by secretion of prostaglandin E2 (PGE2) (Aggarwal &

Pittenger 2005). Interleukin-10 (IL-10) has been suggested as another potential

MSC-derived immunomodulator (Beyth et al. 2005). Interferon-γ (IFN-γ)

production by Th1 type T-cells and NK-cells has been reported to be necessary for

upregulation of indoleamine 2,3-dioxygenase (IDO) in MSCs, leading to

antiproliferative and immunosuppressive effects (Krampera et al. 2006).

Inducible cyclooxygenase 2 (COX2) and inducible nitric oxide synthase (iNOS)

are typical enzymes upregulated in inflammatory conditions. Exposure of

astrocytes to MSCs has been shown to substantially downregulate their

expression (Schafer et al. 2012). Tumor necrosis factor-α (TNFα), interleukin-1β

(IL-1β) and interleukin-6 (IL-6) are key proinflammatory cytokines and

conditioning astrocytes with MSCs significantly attenuates their expression,

together with downregulation of anti-inflammatory IL-10, but not anti-

inflammatory transforming growth factor-β (TGFβ) (Schafer et al. 2012). The

role of TGFβ remains controversial, and some studies have reported that it does

not affect immunomodulation (Beyth et al. 2005).

Systemic inflammation

MSCs have been shown to attenuate systemic inflammatory responses in rodent

models of burn wound and lipopolysaccharide- induced endotoxemia (Yagi et al. 2010a). Systemic immunosuppressive effects of MSCs can be due to decreases in

Page 32: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

30

serum IFN-γ and increases in IL-10 levels (Parekkadan et al. 2008). Lee et al. demonstrated that hMSCs applied in a mouse model of myocardial infarction

were trapped in the lungs and were activated to secrete anti-inflammatory tumor

necrosis factor-stimulated gene 6 protein (TSG-6). These investigators

demonstrated that secretion of this factor suppressed the inflammatory response

to myocardial infarction without a significant effect on cell migration to the

infarcted heart (Lee et al. 2009c).

However, controversial data about the immunomodulatory effects of MSCs

exist. In a study conducted by Hoogduijn et al. in a mouse model, adipose tissue-

derived MSCs targeted the lungs and induced an inflammatory response, with an

increased expression of monocyte chemoattractant protein-1 (MCP-1), IL1-β, and

TNF-α, as well as an increased macrophage accumulation in lung tissue 2 h after

MSC infusion. They also reported increased serum levels of proinflammatory IL-

6 and chemokines CXCL1 (with a CXC motif) and MCP1, demonstrating

systemic immune activation after MSC infusion. MCP1 and TNF-α levels

increased in liver tissue 4 h after MSC infusion without their migration to the

liver. Only a mild effect on expression of the anti-inflammatory cytokines, TGF-

β, IL-4, and IL-10, was observed. However, three days after MSC infusion the

mice developed a milder inflammatory response to lipopolysaccharide, suggesting

that the in vivo immunomodulatory effects of MSCs originated from an

inflammatory response that were induced by the infusion of MSCs, which were

then followed by a phase of reduced immune reactivity (Hoogduijn et al. 2013).

2.2.3 Effects on T-cells

T lymphocytes or T cells constitute the central white blood cell type associated

with cell-mediated immunity. This class of cells is comprised of multiple

subtypes, each of which has their particular role in the immune system. CD4+ T-

helper-cells, the main mediators of adaptive immunity, assist other cells of the

immune system in their processes and are responsible for general activation of the

immune system following presentation with suitable antigens. Subtypes of CD4+

T-helper-cells include Th1-cells, which support cell-mediated immunity, and Th2-

cells, which support antibody responses while inhibiting several functions of

phagocytic cells. CD4+ regulatory T-cells (Treg) maintain self-tolerance and

suppress reactions to weak immunogenic stimuli by inhibiting T-helper-cell

functions. CD8+ T-cells are cytotoxic cells responsible for destroying other cells,

such as virally infected cells or cells that have undergone malignant

Page 33: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

31

transformation, and have been implicated in the rejection of allogeneic transplants

due to the histo-incompatibility between the host and donor organ.

MSCs decrease T-cell proliferation and the percentage of IFN-γ-producing

Th1 T-cells, but do not affect their activation or cytotoxic effector function

(Ramasamy et al. 2008). Some studies have demonstrated that MSCs decrease

IFN-γ secretion by T-cells (Aggarwal & Pittenger 2005, Beyth et al. 2005).

Human MSCs can significantly increase IL-4 production of Th2 cells; MSCs also

increase Th2 differentiation of naïve Th0 T-cells (Aggarwal & Pittenger 2005).

MSCs suppress T-cell proliferation (Bartholomew et al. 2002, Di Nicola et al. 2002, Augello et al. 2005, Krampera et al. 2006), as well as proliferation of both

CD4+ and CD8+ T cells (Di Nicola et al. 2002, Krampera et al. 2006) (Glennie et al. 2005), but some evidence implies stronger suppression of CD8+ than CD4+ T

cells (Krampera et al. 2006). IL-10 has been proposed as a mediator of this effect

and an indirect mechanism has been suggested, whereby T-cell suppression is

established by induction of tolerogenic antigen-presenting cells (APCs) that limit

T-cell activation (Beyth et al. 2005). There is also evidence that T-cell activation

is not suppressed, only proliferation (Glennie et al. 2005). Human MSCs have

been shown to increase the proportion of CD4+CD25+Tregs (Aggarwal & Pittenger

2005).

Direct cell contact is not seen as a requirement for the suppression of T-cell

proliferation (Di Nicola et al. 2002, Krampera et al. 2006). IFN-γ production by

T-cells is reported to be required for MSC IDO upregulation, leading to

antiproliferative and immunosuppressive effects (Krampera et al. 2006). The

exact mechanisms by which MSCs regulate T-cell function remain partially

unsolved, but strong evidence implies suppression of T cell proliferation, thus

explaining the therapeutic results shown in T cell-mediated clinical conditions,

such as reperfusion injury or GvHD.

Effects on natural killer (NK) cells

Human MSCs have been shown to inhibit IFN-γ secretion by NK cells (Aggarwal

& Pittenger 2005) and also their proliferative response to allogeneic stimuli is

diminished (Krampera et al. 2006). IFN-γ production by NK cells has been

reported to be required for MSC IDO-upregulation, leading to antiproliferative

effects (Krampera et al. 2006).

Page 34: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

32

Effects on B cells

MSCs suppress B cell proliferation (Augello et al. 2005), but some studies have

also reported no effect on the B cell proliferative response to allogeneic stimuli

(Krampera et al. 2006). MSC-mediated immunosuppression also affects B cell

proliferation in response to CD40L and IL-4 (Glennie et al. 2005).

Effects on antigen-presenting cells

MSCs reduce TNF-α secretion by activating mature myeloid dendritic cells

(mDCs) and increasing IL-10 secretion of mature plasmacytoid dendritic cells

(pDCs) (Aggarwal & Pittenger 2005). MSCs have been shown to condition DCs

and other APCs to suppress T cell activation (Beyth et al. 2005).

Human MSCs have been shown to suppress initial differentiation of monocyte-

derived DCs produced from CD14+ monocytes reversibly, to suppress moderately

mature monocyte-derived DCs and to inhibit immunostimulation of allogeneic T-

cell proliferation by mature DCs. IL-12 production of DCs was also found to be

suppressed (Jiang et al. 2005).

Variation of immunomodulatory effects by species

The mechanisms of immunomodulation observed are not necessarily directly

translatable to clinical situations because there are differences in the

immunosuppressive mediators that are generated in murine- and human-derived

MSCs (Ren et al. 2009). These findings emphasize the need to carefully consider

preclinical results gained from animal studies on the immunomodulatory

properties of MSCs.

2.3 Clinical applications of MSCs

The main therapeutic potential of MSCs lies in their regenerative and

immunomodulatory capacities. Although the range of theoretical clinical

applications of MSCs is broad, past research has focused on different forms of

ischemic injuries of particular organs, with a heavy emphasis on myocardial

infarction and more recently on ischemic brain injury. The role of MSCs in

ischemic organ injury is not solely due to their regenerative capabilities, as was

initially believed; rather MSC-induced immunomodulation is also recognized as a

Page 35: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

33

primary mode of action in protecting organs against ischemic-induced injuries

(Salem & Thiemermann 2012).

The immunomodulatory applications of MSCs represent captivating

possibilities. These cells can be seen as multifunctional bioreactors with paracrine

and systemic mechanisms of action actions (Ranganath et al. 2012). The clinical

applications of this attribute, in addition to ischemic organ injuries, cover a range

of diseases, which are at least to some extent characterised by a lack of effective

treatment, such as osteogenesis imperfecta or steroid-resistant GvHD (Horwitz et al. 1999, Horwitz et al. 2002, Le Blanc et al. 2008). MSCs and other types of

stem cells therefore offer an experimental and auspicious approach to the

treatment of such conditions of unmet medical need.

2.3.1 Ischemic brain injury and stroke

Ischemic brain injury is most commonly caused by acute thromboembolic events

involving the cerebral circulation (i.e., stroke). A number of factors predispose to

this major cause of morbidity and mortality among Western society, most

importantly atrial fibrillation, hypertension and carotid arterial stenosis stenosis

(Rerkasem & Rothwell 2011, Lager et al. 2014). Another important cause of

ischemic brain injury results from head trauma.

A number of patients also consistently suffer from ischemic brain injury

having an iatrogenic origin. Modern cardiac surgery requires cessation of the

heart’s systolic function and systemic blood flow to perform essential surgical

manoeuvres, such as replacement of heart valves or coronary artery bypass

surgery. This systemic circulatory arrest inadvertently results in ischemic injury to

sensitive organs ― most importantly the brain (Likosky et al. 2003), the kidneys

(Zanardo et al. 1994) and the heart itself ― despite protective actions taken. As

this form of ischemic brain injury is caused by routine clinical procedures, all

precautions taken to prevent this complication are of the utmost importance, with

MSCs representing a promising approach for treating this condition.

Most experimental studies considering MSC transplantation for ischemic

brain injury have utilised a rat MCAO model (Table 1). Better outcome in

behavioural tests compared to controls has been proven in a number of studies

using different transplantation methods (Table 1). A long-term follow-up study of

intravenous autologous mesenchymal stem cell transplantation in patients with

ischemic stroke showed a trend to improvement, with no obvious adverse effects

related to the MSC transplantation (Moniche et al. 2012). Clinical improvement

Page 36: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

34

was correlated with the plasma SDF-1a level at the time of MSC treatment (Lee et al. 2010b). A rodent study with IV MSC transplantation for TBI reported no

improvement in recovery, as the quantity of cell migration was minimal (Harting et al. 2009).

Ischemic brain injury is a particularly interesting application of MSCs due to

the limited effect that the current forms of treatment can offer. The treatment of

stroke mainly consists of thromobolytic agents, which have the disadvantage of

requiring a narrow therapeutic window for treatment. Antihypertensive treatment,

controlling blood glucose levels and medications designed to lower intracranial

pressure comprise the remaining therapeutic options. Ischemic damage to the

brain can be fatal or leave the patient with severe disabilities, with a dependence

on others; hence, developing approaches that lead to even slightest improvements

in treating these conditions can be seen as worthwhile pursuits.

Increasing evidence of the restorative properties of bone-derived stem cells in

treating various disease models is accumulating and ischemic brain injury is no

exception (Table 1-3), although a large gap to pioneer targets of cellular

therapy ― mainly the heart ― exists. Research utilising stem cells in the treatment

of ischemic heart disease and myocardial infarction is advanced, as randomized

controlled trials have already been attempted (Mathiasen et al. 2012, Karantalis et al. 2014).

The brain as a target of cellular therapy differs from many other organs due to

its complex and delicate cellular structure. To date, the possible effects at the

cellular level are seen as a combination of the regenerative, neuroprotective and

immunomodulatory properties of the MSCs

Page 37: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

35

Ta

ble

2. E

xp

eri

me

nta

l in

viv

o s

tud

ies in

ve

sti

ga

tin

g M

SC

th

era

py

fo

r is

ch

em

ic b

rain

in

jury

.

Stu

dy

Mod

el

Rou

te

Gro

ups

Spe

cies

C

ell

num

ber

Tim

ing

Follo

w-

up

Ana

lysi

s O

utco

me

Mig

ratio

n

Bao

et al.

2010

MC

AO

D

irect

inje

ctio

n

hMS

Cs

Con

trol

Rat

5

x 10

5 3

days

28

days

Beh

avio

ural

test

s,

His

topa

thol

ogy,

ELI

SA

, TU

NE

L,

Imm

unoh

isto

chem

istry

Red

uced

infa

rct v

olum

e

Neu

rolo

gic

scor

e im

prov

emen

t

Bor

long

an e

t

al.

2004

MC

AO

D

irect

m

MS

Cs

-5 d

iffer

ent

dose

s

-CB

F or

BB

B

-Pla

cebo

Rat

1,

4, 1

0

or 2

0

x105

Imm

edia

te14

days

Lase

r Dop

pler

, Eva

ns

Blu

e B

BB

ass

ay,

GFP

epi

fluor

esce

nce

mic

rosc

opy,

ELI

SA

Dos

e-de

pend

ent C

BF

and

BB

B

rest

orat

ion,

dos

e-de

pend

ent i

ncre

ase

of g

row

th fa

ctor

s

Che

n et al.

2001

a

MC

AO

D

irect

C

ontro

l

MS

Cs

MS

Cs

+ N

GF

Rat

4

x105

PID

1

14

days

Beh

avio

ural

test

s,

His

topa

thol

ogy,

Imm

unoh

isto

chem

istry

Neu

ral d

iffer

entia

tion

of M

SC

s,

impr

oved

reco

very

Che

n et al.

2001

b

MC

AO

IV

E

arly

/Del

ayed

MS

C/P

lace

bo

Low

/Hig

h

dose

Rat

1

x106

3 x1

06

24h

or

PID

7

7 da

ys

or

35

days

Beh

avio

ural

test

s,

His

topa

thol

ogy,

Imm

unoh

isto

chem

istry

,

CLS

M

Impr

oved

reco

very

with

hig

h do

se

MS

Cs

at P

ID 1

and

PID

7

trans

plan

tatio

n

+

Che

n et al.

2003

MC

AO

IV

hM

SC

s

plac

ebo

Rat

1

x106

1 da

y 14

days

ELI

SA

, His

topa

thol

ogy,

Imm

unoh

isto

chem

istry

Incr

ease

d V

EG

F, in

crea

sed

angi

ogen

esis

Page 38: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

36

Stu

dy

Mod

el

Rou

te

Gro

ups

Spe

cies

C

ell

num

ber

Tim

ing

Follo

w-

up

Ana

lysi

s O

utco

me

Mig

ratio

n

Det

ante

et

al.

2009

MC

AO

IV

M

CA

O

Con

trol

Rat

3

x106

PID

7

PID

7

MR

i, G

amm

a ca

mer

a,

Tiss

ue c

ount

s,

His

topa

thol

ogy

Incr

ease

d br

ain

upta

ke in

MC

AO

grou

p, lu

ng a

ccum

ulat

ion

+

Har

ting

et al.

2009

TBI

IV

TBI

Sha

m

Rat

4

x106

or 1

04 -

106

24h

2 or

14

days

Imm

unoh

isto

chem

istry

,

Infra

red

cell

track

ing,

Beh

avio

ural

test

s

Alm

ost n

o ce

lls in

the

brai

n at

PID

14,

no im

prov

ed re

cove

ry, p

rom

inen

t lun

g

accu

mul

atio

n, s

mal

l num

ber o

f cel

ls

reac

hing

arte

rial c

ircul

atio

n

Kei

mpe

ma

E. e

t al.

2009

MC

AO

IA

M

SC

s

Pla

cebo

Rat

1

x 10

6 2h

P

ID 1

4 H

isto

path

olog

y,

Imm

unoh

isto

chem

istry

CLS

M

Red

uced

lesi

ons,

onl

y a

few

cel

ls

dete

cted

afte

r 2 w

eeks

+

Koh

et al.

2008

MC

AO

D

irect

hU

C-M

SC

s

Con

trol

Rat

6

x 10

5 P

ID 1

4 P

ID 3

5 B

ehav

iour

al te

sts,

Imm

unoh

isto

chem

istry

,

MR

i

Impr

oved

reco

very

, red

uced

lesi

on

size

, neu

rotro

phic

fact

or e

xpre

ssio

n

Lee

et al.

2010

a

Uni

late

ral

caro

tid

occl

usio

n

IA

(intra

-

card

iac)

hMS

Cs

or

plac

ebo

Rat

,

neon

atal

1 x

106

72h

PID

40

Beh

avio

ural

test

s,

His

topa

thol

ogy

Impr

oved

reco

very

, red

uced

lesi

on

size

Par

k et al.

2011

TBI

IV

Trau

ma

Con

trol

Rat

1

x106

24h

24h

+

4h

Gam

ma

cam

era,

Tiss

ue c

ount

s

Incr

ease

d br

ain

upta

ke in

trau

ma

grou

p by

gam

ma

coun

ter,

gam

ma

cam

era

upta

ke in

diffe

rent

She

n et al.

2006

MC

AO

IA

M

SC

s

Pla

cebo

Rat

2

x 10

6 P

ID 1

P

ID 2

8 B

ehav

iour

al te

sts,

His

topa

thol

ogy,

Imm

unoh

isto

chem

istry

Impr

oved

reco

very

, inc

reas

ed O

PC

coun

t and

mar

kers

rela

ted

to a

xon

and

mye

lin re

mod

ellin

g

Page 39: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

37

Stu

dy

Mod

el

Rou

te

Gro

ups

Spe

cies

C

ell

num

ber

Tim

ing

Follo

w-

up

Ana

lysi

s O

utco

me

Mig

ratio

n

de

Vas

conc

elos

Dos

San

tos

et al.

2010

Foca

l

corti

cal

isch

emia

IV

BM

-MN

Cs

MS

Cs

Pla

cebo

Rat

B

M-

MN

Cs:

3 x1

07

MS

Cs:

3 x1

06

1, 7

, 14

or

30 d

ays

77

days

Beh

avio

ural

test

s,

Imm

unob

lotti

ng

Impr

oved

reco

very

whe

n B

M-M

NC

s

trans

plan

ted

at 1

or 7

day

pos

t-op,

MS

Cs

at 1

day

, BM

-MN

Cs

and

MS

Cs

equa

l

Page 40: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

38

Ta

ble

3. E

xp

eri

me

nta

l in

viv

o s

tud

ies in

ve

sti

ga

tin

g B

M-M

NC

th

era

py

fo

r is

ch

em

ic b

rain

in

jury

.

Stu

dy

Mod

el

Rou

teG

roup

s S

peci

esC

ell n

umbe

r Ti

min

g Fo

llow

-

up

Ana

lysi

s O

utco

me

Mig

ratio

n

Fujit

a et al.

2010

BC

AS

BC

AS

-BM

-MN

Cs

Sha

m

Mou

se

5 x1

06 24

h 30

day

s La

ser s

peck

le

perfu

sion

imag

ing

Aug

men

ted

cere

bral

blo

od

flow

, les

s w

hite

mat

ter

dam

age,

incr

ease

d V

EG

F

Iihos

hi e

t a

l.

2004

MC

AO

IV

B

M-M

NC

s: 3

-

72h

Pla

cebo

: 3-

72h

Sha

m

Rat

1

x107

3, 6

, 12,

24

or 7

2h

B

ehav

iora

l tes

ts,

His

topa

thol

ogy,

CLS

M

Red

uced

lesi

on in

all

grou

ps,

earli

er ti

min

g m

ost e

ffect

ive,

impr

oved

reco

very

+

Kam

iya

et

al.

2008

MC

AO

IV

IA

IV, I

A

Pla

cebo

Rat

1

x107

Imm

edia

te

7 da

ys

His

topa

thol

ogy,

Beh

avio

ral t

ests

Red

uced

infa

rct v

olum

e in

IV

and

IA g

roup

s, im

prov

ed

reco

very

in IA

gro

up

de

Vas

conc

elos

Dos

San

tos

et al.

2010

Foca

l

corti

cal

isch

emia

IV

BM

-MN

Cs

MS

C

Pla

cebo

Rat

B

M-M

NC

s: 3

x 10

7

MS

Cs:

3 x

106

1, 7

, 14

or 3

0

days

77 d

ays

Beh

avio

ral t

ests

,

Imm

unob

lotti

ng

Impr

oved

reco

very

whe

n B

M-

MN

Cs

trans

plan

ted

at P

ID 1

or 7

, MS

Cs

at 1

day

BM

-

MN

Cs

and

MS

Cs

equa

l.

Page 41: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

39

Ta

ble

4. C

lin

ica

l stu

die

s i

nv

esti

gati

ng

BM

-MN

C o

r M

SC

th

era

py

fo

r is

ch

em

ic b

rain

in

jury

.

Stu

dy

Dis

ease

Line

age

Rou

teG

roup

s P

atie

nts

Sou

rce

Cel

l

num

ber

Tim

ing

Follo

w-

up

Ana

lysi

s O

utco

me

Mig

ratio

n

Bar

bosa

da

Fons

eca

et

al.

2009

MC

A

Stro

ke

BM

-

MN

Cs

IA

No

cont

rol

n =

1 A

utol

ogou

s5

x108

PID

67

2, 2

4,

48h

Gam

ma

cam

era

SP

EC

T

Cel

l acc

umul

atio

n in

lesi

on, n

o

adve

rse

effe

cts

+

Bar

bosa

da

Fons

eca

et

al.

2010

MC

A

Stro

ke

BM

-

MN

Cs

IA

No

cont

rol

n =

6 A

utol

ogou

s1.

25 x

108

to 5

x 1

08

< P

ID

90

120

days

Gam

ma

cam

era,

SP

EC

T

Cel

l acc

umul

atio

n in

lesi

on, n

o

adve

rse

effe

cts

+

Cor

rea

et

al.

2007

Stro

ke

BM

-

MN

Cs

IA

No

cont

rol

n =

1 A

utol

ogou

s3.

0 x

107

PID

9

4 mon

ths

SP

EC

T C

ell a

ccum

ulat

ion

in le

sion

at 8

h,

no a

dver

se e

ffect

s

+

Cox

et al.

2011

TBI i

n

child

ren

BM

-

MN

Cs

IV

No

cont

rol

A

utol

ogou

s6

x106 /k

g B

efor

e

PID

2

6 mon

ths

PE

LOD

scor

ing,

Neu

rolo

gic

reco

very

, MR

i

No

adve

rse

effe

cts,

neu

rolo

gic

scor

e im

prov

ed

n/a

Mon

iche

et

al.

2012

MC

A

Stro

ke

BM

-

MN

Cs

IA

BM

-

MN

Cs

Con

trol

n =

10

n =

10

Aut

olog

ous

1.59

x 1

08

(mea

n)

PID

6.4

(±1.

3)

6 mon

ths

GM

CS

F,

BN

GF,

MR

i

Neu

rolo

gy

Impr

oved

reco

very

Page 42: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

40

2.3.2 Other indications

Although ischemic brain injury is an interesting application for MSCs, several

other indications have shown promising results with cellular therapy. Some of the

indications offer a possibility for either direct or systemic transplantation of the

cells, but only systemic transplantation is an option with certain diseases.

Myocardial infarction

Myocardial infarction resulting from coronary arterial disease is the single most

important cause of mortality and morbidity in western countries. The therapeutic

potential of MSCs is well documented in the treatment of myocardial infarction in

experimental models, with the number of clinical trials growing steadily.

Intravascular delivery, either intravenously or via the intracoronary route, is

shown to reduce the infarct volume and preserve the ejection fraction of the heart,

thus protecting the vital function of the heart while maintaining the integrity of

circulatory homeostasis (Chou et al. 2014). Due to the vast number of cases and

the potentially lethal nature of ischemic cardiac events, MSCs represent a

promising approach if they can be applied to the prevention of ischemic damage

or the regeneration of previously injured myocardium.

Osteogenesis imperfecta

Osteogenesis imperfecta (OI) is a rare hereditary disorder of bone development

characterized by fragile bony structures that are prone to fractures. The

underlying defect is a mutation in one of the two genes encoding type I collagen,

the primary structural protein of bone. The severity of the disease varies. Some

variants of OI cause inevitable perinatal morbidity, whereas patients carrying

milder forms of the disorder survive into adulthood, but suffer from pathologic

fractures and skeletal deformities. No curative treatment currently exists for the

disorder. Clinical case reports with MSCs have offered encouraging results in the

treatment of OI, showing trends toward increased bone mineral content and

decreased occurrence of fractures (Horwitz et al. 1999), and even evidence of

accelerated growth (Horwitz et al. 2002). Graft-versus-host-disease

Organ transplantation is an advanced procedure for treating various severe

clinical conditions and is usually performed as a last resort due to the complexity

Page 43: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

41

of the required surgical operation, the shortage of suitable organ donors and the

inherent problems associated with allogeneic organ transplantation. The

postoperative complications consist of organ rejection by the recipient’s immune

system and GvHD, in which the allograft starts to attack the host. Both conditions

have severe consequences, including loss of the allograft or even host mortality in

severe GvHD cases. MSCs have emerged in the treatment of steroid-resistant

GvHD following HSC transplantation. A number of patients were reported to

completely respond in a clinical Phase II trial, as reported by LeBlanc et al. (Le

Blanc et al. 2008).

Renal transplant rejection

Renal failure and ultimately end-stage renal disease is caused by a variety of

aetiologic clinical conditions, conditions that are both acquired, such as diabetes,

or inherited (e.g. polycystic kidney disease). The loss of renal function requires

patients to undergo dialysis multiple times per week to filter excess urea and

creatinine from the blood. Renal allograft transplantation replaces the need for

dialysis and significantly decreases the treatment dependency of the patient, but

allograft rejection is a common complication, with a risk of graft loss and patient

mortality.

A Phase I clinical trial reported the usage of autologous MSCs for renal

transplant rejection as being safe; potential therapeutic effects were observed, but

possible mechanisms behind this effect remain unknown (Reinders et al. 2013). A

randomised controlled trial reported that autologous MSCs, compared with anti-

IL-2 receptor antibody induction therapy, resulted in a lower incidence of acute

rejection and a better outcome (Tan et al. 2012).

Other indications

A constantly growing number of MSC clinical trials are registered to treat various

other disorders in addition to those previously mentioned. Mesenchymal stem

cells have been shown to migrate to damaged lung tissue (Lee et al. 2009a, Lee et al. 2009b). A Phase I/II double-blinded, randomized, placebo-controlled clinical

trial utilizing locally injected MSCs showed positive trends among ankle-brachial

pressure index and ankle pressure in patients suffering from critical lower limb

ischemia (Gupta et al. 2013). Universal arterial stenosis is a common

manifestation of atherosclerotic arterial disease, often leading ultimately to

Page 44: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

42

amputations of lower limbs, resulting in severe disability. Use of MSCs have also

emerged in the treatment of liver cirrhosis patients, who showed positive effects

in a Phase I/II clinical trial (Kharaziha et al. 2009). Huntington’s disease

(Bantubungi et al. 2008) and tissue engineering of artificial bones (Petite et al. 2000) are yet additional examples of the range of possible MSC applications.

2.4 Intravascular transplantation of bone marrow-derived cells

The optimal method to deliver stem cells in vivo is a matter of debate. In

preclinical animal studies, numerous delivery routes have been attempted,

including direct injection into the target area and intravascular delivery (either via

intravenous or intra-arterial routes).

For direct injection, it can be theoretically assumed that this route would

result in the delivery of a large effective cell dose to the target area. However,

direct injection (to the heart or the brain for example) is a considerably invasive

procedure with inherent risks and no direct evidence supports such an approach

for stem cell transplantation for these indications. Intravascular transplantation

could be considered a more clinically relevant approach. Intravenous

transplantation is a particularly appealing method due to its simplicity, non-

invasiveness and assumed safety. In addition, this route would naturally be the

method of choice in clinical conditions, where a systemic approach would be

reasonable, such as in GvHD.

Intra-arterial transplantation should theoretically overcome the issues

associated with using an IV route to a certain extent. Delivering the cells through

an arterial route would allow the cells to avoid the first passage through the

pulmonary circulation. The cells could either be administered systemically to the

arterial circulation via large central arteries to allow natural distribution in the

vascular compartment or they could be targeted very specifically to the lesion

using modern catheterisation techniques. Catheterisation would allow the cells to

be administered directly to coronary arteries or cerebral arteries, thereby allowing

a precise targeting of MSC therapy to the subject.

Preclinical trials with animal models offer the opportunity to explore more

invasive transplantation methods other than intravascular routes. However, issues

of safety are critical when dealing with actual patients and it is of the utmost

importance that the risks of the transplantation procedure do not exceed the

potential therapeutic benefits gained with the transplantation.

Page 45: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

43

2.4.1 The role of direct transplantation

In addition to intravascular routes, direct transplantation could also offer some

advantages in certain clinical settings, as their paracrine effects can take place at

the site of injury; an example for such indication would be intramyocardial

injection during cardiac surgery (Lehtinen et al. 2014). Direct intracranial

transplantation has shown positive effects in animal studies (Chen et al. 2001a).

When comparing direct BM-MNC transplantation and IV transplantation

effectiveness in a rat spinal cord lesion model, direct injection resulted in more

potent remyelination with smaller cell doses, but the same effect could also be

achieved with larger IV cell doses (Inoue et al. 2003). Direct intramyocardial

transplantation has been shown to improve lesional homing in a porcine MI

model compared with intracoronary transplantation (Makela et al. 2009). Some

other possible indications for local transplantation could be in the treatment of

Huntington’s disease (Bantubungi et al. 2008) and in the tissue engineering of

artificial bones (Petite et al. 2000). The potential benefits naturally come at the

expense of considerably more invasive approaches.

Novel approaches of direct transplantation include different cell

encapsulation techniques, whereby therapeutic cells are transplanted within

biocompatible-carrying materials such as gels. Encapsulation aims to enhance the

survival and retention of cells in the target area. Ideally, the encapsulation

materials protect the cells from mechanical stimuli, but the porosity of the

material allows the secretome to exert its paracrine function (Goren et al. 2010,

Levit et al. 2013).

Another factor to be considered with direct transplantation relates to

observations of possible therapeutic benefits due to the accumulation of cells in

the lung (Lee et al. 2009c), raising the question as to whether some of the

systemic therapeutic characteristics are lost due to local transplantation.

Presumably, MSCs that are directly transplanted to the target of therapy do not

reach the systemic circulation in relatively large numbers (Makela et al. 2009),

thus resulting in lung deposition. In addition, some of the systemic secretome-

based effects could theoretically even arise from migration to certain organs other

than the lung, although there is currently no evidence supporting this hypothesis.

Inoue et al. (2003) compared BM-MNC dose on transplantation-dependent

remyelination in a rat spinal cord injury model. Although direct transplantation

had a better remyelination rate, comparable results could also be achieved with IV

transplantation; the equal dose reported in the study was 1 × 107 autologous bone

Page 46: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

44

marrow cells IV and direct intraspinal injection of 1 × 105 cells (Inoue et al. 2003). This finding would favour the use of an intravascular transplantation route,

as no neurosurgical intervention for cell transplantation would be needed and

comparable results could still be achieved.

2.4.2 Passive biodistribution after intravascular transplantation

Intravenous transplantation can be executed by cannulating peripheral or central

vessels. Intravenously transplanted substances, such as bone marrow-derived cells

diluted with a solution, circulate through the venous system, passing through the

right atrium and ventricle of the heart to the pulmonary circulation, inevitably

encountering the lung capillaries. After passing through the lung vasculature, the

cells reach the systemic circulation after they are ejected from the left ventricle.

The arterial circulation begins from the left ventricle, which ejects the blood

volume through the whole systemic vasculature, reaching arteries of diminishing

calibre all the way down to the peripheral capillaries. After passing through the

capillary level, the blood passes again through peripheral veins, only to reach the

heart once again.

If mechanisms of active migration are not taken under consideration, it could

be assumed that the cells are passively targeted to various organs with such

distribution as the blood stream follows. Certain organs (i.e, the brain and the

kidneys) use the majority of the heart’s minute volume and this proportion is not

necessarily commensurate to the organs weight. Circulating blood volume

distribution is also greatly affected by the systemic state of the body, being

regulated by constriction or relaxation of the vessel walls.

2.4.3 Active biodistribution by chemotaxis and migration

A common observation noted in various studies is that systemically administered

MSCs are targeted to injured tissues. Acute inflammation due to organ damage

increases local circulation, but the results found suggest an active migration of the

transplanted cells.

The production of SDF–1 (Stromal cell derived factor–1α), is a chemokine,

which production increases as a result of damage to cellular DNA (Ponomaryov et al. 2000). The CXCR4 receptor binds to SDF-1 and increases stem cell migration

to the SDF-1-secreting tissue (Bhakta et al. 2006). A lung contusion model

performed with rats showed an increased bone marrow-derived cell migration to

Page 47: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

45

the damaged area after SDF-1 injection and resulted in improved healing

(Hannoush et al. 2011). It has been demonstrated with a myocardial infarction

model that SDF-1 enhances stem cell homing to the damaged area, where SDF-1

is forcibly overexpressed in myocardium, but not in the absence of myocardial

infarction (Abbott et al. 2004).

Controversial mechanisms of tissue migration have also been suggested. The

role of CXCR4, as well as integrin α4 in MSC engraftment in a MI model has

been suggested to be of less importance, whereas integrin β1 has been shown to

facilitate migration and engraftment in MI (Ip et al. 2007). A broader spectrum of

chemokines and their receptors has been hypothesised to mediate MSC tissue

migration, with CXCL8 (interleukin-8) having also been verified to increase

migration (Ringe et al. 2007). The impact of SDF-1 on MSC migration has been

questioned in the study of Ries et al. 2007. Instead, the inflammatory cytokines

TGF-β1, IL-1β, and TNF-α function by acting as chemoattractants for MSCs,

augmenting their invasive capacity by up-regulation of MMP-2, MT1-MMP,

and/or MMP-9 activity in these cells (Ries et al. 2007). CD44 interaction with

extracellular hyaluronic acid has been reported to facilitate MSC migration (Zhu et al. 2006).

Bone marrow stem cell-specific mechanisms of migration are not yet

completely resolved, but similar mechanisms are suggested to be involved, as

seen with white blood cells. It has been shown that MSCs are capable of

coordinated rolling behaviour and adhesion to endothelial cells, as they bind to P-

selectin and VCAM-1, thus fulfilling the basic requirements of extravasation to

various organs (Ruster et al. 2006). Cell adhesion of MSCs mediated by the

adhesion molecules ICAM-1 and VCAM-1 is considered critical for mediating

the immunosuppressive effects of MSCs (Ren et al. 2010).

2.4.4 Biodistribution tracking methods

In order to assess how the transplanted MSCs distribute among the vasculature

and engraft into organs, studies must be conducted using methods that allow the

monitoring of the fate of the cells. An ideal tracking method would accurately

reveal global cell deposition throughout the whole body, the method would not

interfere with the cells naïve biological characteristics and therapeutic potential

and the method should be safe to the subject and be as minimally invasive as

possible to avoid introducing effects of entry trauma, possibly biasing cell

distribution. Preclinical animal studies can compromise certain safety measures if

Page 48: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

46

more precise information of the cellular fate is gained in return; however,

methods applied with human subjects must be safe for the patient.

Radionuclide imaging

Radionuclide imaging is a widely used method in which a radioactive substance is

injected into the subject and tracked with equipment able to detect the emitted

radiation, followed by reconstruction of images from the data. In a variety of

clinical applications a common method is to intravascularly inject the substance,

such as glucose or leukocytes that have been labelled with a radioactive isotope.

The clinical applications cover a large range of different studies, including

leukocyte scintigraphy or metabolic mapping used in cancer diagnostics.

Different imaging methods exist; generally, the more advanced methods have

higher spatial resolution and accuracy, but study costs are significantly higher and

study execution more elaborate. Scintigraphy is a basic form of radionuclide

imaging in which the gamma radiation emitted from the radiotracer is detected by

a gamma camera, forming two-dimensional planar images from the subject. In

single photon emission tomography (SPECT), the gamma-radiation is measured

from multiple directions, thereby allowing reconstruction of three-dimensional

images. SPECT possesses the ability to combine the acquired imaging data with

computed tomography (CT) images taken from the same subject. The

combination of these two imaging methods is called SPECT/CT. Positron

emitting tomography (PET) allows more precise spatial resolution; PET-scanners

detect pairs of gamma rays emitted indirectly by a positron-emitting radionuclide

and it can also be combined with other forms of tomographic imaging, such as

CT.

Radionuclide imaging provides the possibility to dynamically assess the fate

of the transplanted therapeutic cells (Kraitchman et al. 2005). Experimental

radionuclide imaging methods are adopted from clinical practice, where the

application most transferable to stem cell imaging is the labelling of leukocytes.

This procedure, combined with imaging methods, is primarily used in whole body

mapping of infection foci. The most feasible radiotracers for MSC labelling are

technetium99m-hydroxymethylpropylene amine oxime (Tc99m-HMPAO) or indium-

111 oxine. The isotope half-life of these tracers are 6.01 hours and 2.80 days,

respectively.

Radionuclide imaging is considered to be the only method for MSC

biodistribution mapping available and provides the opportunity to acquire global

Page 49: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

47

body biodistribution imaging data. The disadvantage of radionuclide methods is

the limited half-life of the radiotracer, resulting in diminishing amounts of emitted

gamma-radiation, which restricts the monitoring of the transplanted MSCs.

Additionally, it is possible that radiolabelling of the transplanted cells could affect

their biological characteristics or compromise their viability. In addition, this

method naturally exposes the target to a certain dose of radiation, but the dose can

generally be considered tolerable as the exposure resulting from the radiotracers

would presumably be the same as that applied in daily clinical practice.

Tissue counts

In addition to actual radionuclide imaging modalities, labelling the transplanted

MSCs with radiotracer isotopes enables the quantitation of tissue counts. Organic

material, such as biopsies from target organs or even whole organs in the case of

small animal models, can be placed in a gamma detector. The counts measured

can be normalised to the weight of the sample, thus making samples obtained

from various organs comparable.

The method can be considered very specific, as the samples are visually taken

from the target organ, thus avoiding bias of scattering radiation from surrounding

organs or limitations set by imaging resolution. An obvious weakness of the

method is that biopsies have to be relatively large, hence limiting the use for post-

mortem analysis and naturally preclinical animal experiments. This means that

using tissue counts, MSC distribution can only be assessed at one time point in a

single subject. Another possibly biasing factor is that if biopsies are obtained from

a large organ, they may not represent the whole organ’s MSC density if the cells

are unevenly distributed throughout the organ. Assessment of overall distribution

of the transplanted cells is challenging with tissue counts, as representative tissue

samples would have to be obtained from all organs. However, tissue samples are a

feasible tracking method if MSC quantification is limited to selected targets and

overall distribution is not of a concern.

Magnetic resonance imaging

Magnetic resonance imaging (MRi) is an imaging technique that uses the

application of a strong magnetic field. Imaging equipment detects radiofrequency

signals emitted by excited hydrogen atoms in the subject’s body. The resulting

Page 50: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

48

images are accurate and the MRi does not expose the subject to any ionizing

radiation.

MRi can be applied in the assessment of the biodistribution of MSCs after

labelling of the cells with radio-contrasting agents. Super paramagnetic iron oxide

labelling has been used in MSC studies, which makes the transplanted cells

detectable from MRi images. MRi is not as sensitive as radionuclide imaging

methods in detecting the transplanted cells, as reported in a comparative analysis

in a canine MI model. In this study, MRi failed to detect ferumoxine- and

indium-labelled IV transplanted MSCs in the subject’s myocardium, which were

detectable by SPECT/CT (Kraitchman et al. 2005). This questions the feasibility

of using MRi as an overall MSC biodistribution tracking method, as even a small

number of transplanted cells migrating to the target organ could possess

therapeutic significance, as described in a model of ischemic brain injury

(Keimpema E. et al. 2009). However, even with limited sensitivity to detect the

transplanted cells, MRi offers a range of features not available with other tracking

methods, such as assessment of myocardial ischemia and different forms of

functional brain imaging.

Optical tracking methods

Labelling the transplanted cells with self-illuminating substances, such as

luciferase, offers a method of non-invasive imaging. Self-illuminating proteins

can be introduced into target cells by viral vectors. The transplanted cells can then

be detected in a bioluminescence chamber with an ultrasensitive camera. The

method is non-invasive and allows longitudinal study setups (Wang et al. 2009).

The transplanted MSCs can be labelled with histologic stains, which allow

them to be detected in histopathologic samples (e.g. organ biopsies). This makes

the transplanted cells detectable in microscopic samples. However, this method is

not feasible in the quantification of the cells and is limited only to confirming the

presence of the cells. Assessment of overall biodistribution encounters the same

challenges as tissue count biopsies, as representative samples are hard to obtain

for comparison between organs. The value of histopathology and

immunohistochemistry methods lies in their ability to evaluate the cellular fate,

differentiation, survival and even functionality of the transplanted cells. Possible

labels that allow MSC tracking include β-galactosidase (β-gal), Green

Fluorescence Protein (GFP), chloromethyl-dialkylcarbocyanine (DiI), Hoechst

dyes, 4',6-diamidino-2-phenylindole (DAPI), 5-bromo-2'-deoxyuridine (BrdU)

Page 51: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

49

and 5-ethynyl-2’-deoxyuridine (EdU) (Lin et al. 2013). Another novel approach

for MSC labelling is the use of quantum dots, which can be applied in the

imaging of intra- and inter-cellular communication (Pietila et al. 2013).

Polymerase chain reaction

DNA of the transplanted cells can be detected from tissue samples by application

of the polymerase chain reaction (PCR). This method can be applied in

xenogeneic transplantation. Usually the transplanted cells are of human origin,

thus rendering the possibility to isolate human DNA from the experimental

animal’s tissues. PCR allows the potential to confirm the presence of the

transplanted cells after prolonged follow-up (Meyerrose et al. 2007).

2.4.5 Acute phase biodistribution

A common pattern of bone marrow-derived cell biodistribution after intravascular

transplantation consists of strong early lung deposition (Gao et al. 2001, Barbash et al. 2003, Detante et al. 2009, Barbosa da Fonseca et al. 2010, Kraitchman et al. 2005, Allers 2011, Vasconcelos-dos-Santos et al. 2012). The rest of the cells

localise mostly in the liver and spleen (Gao et al. 2001, Barbash et al. 2003,

Kraitchman et al. 2005, Detante et al. 2009, Barbosa da Fonseca et al. 2010,

Vasconcelos-dos-Santos et al. 2012), followed by kidneys (Table 4).

Biodistribution is considered as a dynamic effect and lung clearance occurs

during the first few days. This redistribution results in a proportional increase of

cells deposited in the liver, spleen, kidneys (Chin et al. 2003, Kraitchman et al. 2005) and spine (Kraitchman et al. 2005). Redistribution appears to occur

relatively quickly following transplantation, with an initial 80 % of cell

entrapment for the BM-MSCs decreasing to 40 % after 12h (Nystedt et al. 2013).

Thus, it could be hypothetically estimated that the “half-life” of MSC lung

entrapment is roughly 12 hours.

Unfortunately, little is known about the naïve biodistribution of the

transplanted MSCs, as nearly all the studies employed some form of target organ

(Table 4). Usually ischemic organ damage is induced by experimental methods in

preclinical studies (e.g. ligation of coronary vessels or MCAO occlusion) or, in

the case of human studies, organ damage has already occurred as a result of a

natural disease process, such as atherosclerosis or embolisation. Ischemic injury

of a major organ alters the inflammatory state of the entire system, which

Page 52: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

50

presumably affects MSC immunologic characteristics, thereby causing MSC

migration to the injured organ as observed previously (Abbott et al. 2004, Bhakta et al. 2006, Hannoush et al. 2011). It is currently unknown how the organ injury

and resulting alterations in the immune system affect the MSC deposition in

organs other than the injured one. Devine et al. reported a study in which two

unconditioned baboons received autologous and allogeneic MSCs. These

transplanted cells could be detected in significant amounts in gastrointestinal

tissues, as well as in kidneys, lungs, liver, thymus, and skin, as assessed by real-

time PCR (Devine et al. 2003).

2.4.6 Long-term biodistribution

Although, the studies offer somewhat comprehensive insights into the acute

biodistribution of the transplanted cells, the long-term fate of the transplanted

cells remains partially unknown. Radionuclide methods, which potentially offer

the most accurate way of assessing overall distribution, cannot be used to observe

long-term engraftment due to the limited half-life of the radioactive isotopes.

Some studies have attempted to elucidate long-term engraftment. It appears that

adipose tissue-derived hMSCs persist in the tissues of a murine model for up to

75 days and can be found mainly in the lungs and the spleen, regardless of the

route of administration, while retaining their multilineage potential and

clonogenic capacity (Meyerrose et al. 2007). IV or intramuscularly transplanted

human adipose tissue-derived mesenchymal stem cells showed long-term

engraftment to the liver in a murine model after an 8-month follow-up, as

assessed by bioluminescence imaging, and no tumour formation was observed

(Vilalta et al. 2008). Interestingly, human MSCs have shown long-term deposit

into lungs when intravenously transplanted into unconditioned mice, as human

DNA could be detected 4 months after transplantation (Allers et al. 2004). It can

thus be concluded that the transplanted cells may survive for prolonged periods

after transplantation and engrafting into multiple organs.

2.5 Challenges of intravascular MSC transplantation

Although intravascular transplantation is a promising method of delivering the

therapeutic cells, certain complications related to the procedure have to be taken

under consideration. The transplantation can either be unsuccessful or even

posess potential hazard to the patient.

Page 53: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

51

2.5.1 Periprocedural damage to MSCs

The bone marrow-derived cell’s natural environment (namely the bone marrow) is

characterized by relatively low oxygen content, high cell density and consistency

varying from solid trabecular bone to highly viscose and fatty bone marrow. The

MSCs are located in perivascular niches, where they constantly interact and co-

exist with other types of cells, such as HSCs and macrophages.

Compared to the natural environment of MSCs, the intravascularly

transplanted cells are subjected to various circumstances that can be considered

unnatural and injurious. These conditions may lead to cellular death or damage of

the cells, which could affect their therapeutic capacity due to mechanical and

biochemical damage.

Mechanical damage to MSCs can be caused by various reasons. Cells are

usually aspirated into syringes and injected from them when they are harvested

from the bone marrow, handled and relocated in cultivation procedures or

transplanted into target organisms. MSCs have a strong tendency to attach onto

materials, such as cultivation plates, which requires the use of detaching

substances and mechanical detachment. Mechanical forces caused by the

circulatory system can also expose the cells to physical damage, especially in the

arterial circulation as the cells are forcefully pushed by the blood stream through

the small-diameter capillary structures of the lungs, liver and various other

organs.

The MSCs are suspended into various solutions during the isolation and

cultivation process, which also can be considered as non-physiological conditions

compared to their natural environment. Additionally, the intravascular

transplantation of the cells cultivated on culture dishes leads to a drastic increase

of surrounding oxygen content that could cause chemical damage. The lack of

maintenance cells, namely macrophages, in culture dishes prevents the clearance

of unfit or damaged cells from the culture population. Compared to signal-

induced activation and migration of MSCs from the bone marrow to the systemic

circulation, mechanical harvesting, culturing and direct injection into target

tissues or the circulation truly challenges the survival potential of the MSC to

elude cellular death due to damage, as well as the retaining of their therapeutic

characteristics.

Page 54: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

52

Ta

ble

5. S

tud

ies

in

ve

sti

ga

tin

g b

on

e m

arr

ow

de

riv

ed

ce

ll a

cu

te b

iod

istr

ibu

tio

n.

Stu

dy

Mod

el

Spe

cies

Trac

king

C

ells

N

umbe

r R

oute

Tim

ing

Qua

ntita

tion

Follo

w-

up

Gro

ups

Bra

inLu

ng

Live

rS

plee

n K

idne

yH

eart

Not

e

Alle

rs e

t a

l.

2004

Hea

lthy

Mou

se

Tc99

m-

HM

PA

O

PC

R

hMS

Cs

0.2–

2x10

5 IV

n/

a W

hole

bod

y

scan

, PC

R

15m

in,

3h, 2

4h

+++

+++

+

Bar

bash

et

al.

2003

MI

Rat

Tc

99m

-

HM

PA

O

MS

Cs

4x10

6 IV

, IA

PID

2,

10 o

r 14

Imag

ing

afte

r 4h

Who

le b

ody

scan

Tis

sue

coun

ts

IV

LV

- -

+++ +

+ ++

+ ++

+ +

+ +

Bar

bosa

da

Fons

eca

et

al.

2009

Stro

ke

Hum

an

Tc99

m-

HM

PA

O

BM

-MN

Cs

5 x1

08 IA

SP

EC

T 2h

, 24h

,

48h

++

+

++

++

-

Chi

n e

t a

l. 20

03

MI

Pig

11

1In

oxin

e

MS

Cs

n/a

IV

n/a

SP

EC

T

Gam

ma

cam

era

Tiss

ue

coun

ts

++

+/++

+/++

+/++

+

-

Det

ante

et

al.

2009

MC

AO

R

at

Tc99

m-

HM

PA

O

hMS

Cs

3 x1

06 IV

2h

or

20h

MC

AO

Con

trol

+ ++

+ +/

- +

+

Kra

itchm

an e

t

al.

2005

MI

Dog

MS

Cs,

allo

gene

ic

1.13

x108

IV

72h

SP

EC

T P

ID

1,2,

5

and

8

MI

Con

trol

n/a

+++/

+++/

+++/

++

+/++

+

Mitk

ari e

t a

l.

2013

MC

AO

R

at

Indi

um-

111-

oxin

e

MS

Cs,

allo

gene

ic

1.1

x 10

6 or

0.5

x 10

6

IA

24h

SP

EC

T 30

min

to 2

4h

MC

AO

/

Sha

m +

mod

ified

/

nonm

odifi

es

cells

+ +

+++

+++

++

+ Lu

ng a

ctiv

ity

decr

ease

d

over

tim

e

Page 55: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

53

Stu

dy

Mod

el

Spe

cies

Trac

king

C

ells

N

umbe

r R

oute

Tim

ing

Qua

ntita

tion

Follo

w-

up

Gro

ups

Bra

inLu

ng

Live

rS

plee

n K

idne

yH

eart

Not

e

Vas

conc

elos

-

dos-

San

tos

et

al.

2012

Stro

ke

Rat

Tc

99m

- B

MM

Cs

3 x

107

IV, I

A

24h

Gam

ma

coun

ter,

Tiss

ue c

ount

s

S

troke

/

cont

rol +

IV/

IA

+/-

++/-

+++

+ ++

-

Lung

act

ivity

decr

ease

d

over

tim

e

Page 56: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

54

2.5.2 Pulmonary adhesion and the lung barrier

Multiple studies have reported robust lung accumulation of the transplanted cells.

(Gao et al. 2001, Barbash et al. 2003, Detante et al. 2009, Barbosa da Fonseca et al. 2010, Kraitchman et al. 2005, Allers 2011, Vasconcelos-dos-Santos et al. 2012) This phenomenon is considered to be potentially harmful, as pulmonary

adhesion of the cells could lead to a lesser proportion of the cells reaching the

arterial circulation and their intended therapeutic target, hence hypothetically

attenuating their effectiveness and migration to other organs (Barbash et al. 2003,

Fischer et al. 2009, Freyman et al. 2006, Lee et al. 2009c). Lung entrapment is

especially observed after IV transplantation (Allers et al. 2004, Barbash et al. 2003, Barbosa da Fonseca et al. 2010, Detante et al. 2009, Gao et al. 2001,

Kraitchman et al. 2005, Makela et al. 2013, Vasconcelos-dos-Santos et al. 2012)

and it appears that the majority of the transplanted cells first target the lungs in

large numbers. Intravenous transplantation of MSCs was even shown to cause

lethal pulmonary emboli in preclinical studies (Lee et al. 2009d). Schrepfer et al. named this prominent lung deposition as lung entrapment (Schrepfer et al. 2007).

Physical lung entrapment purely due to the size of the transplanted cells has

been suggested by some recent studies. Fischer et al. observed a 30-fold greater

lung accumulation in MSC transplantation compared to BM-MNCs and based on

their larger diameter (5 to 8 µm cell size vs 13 to 19 µm, respectively), MSCs do

not fit through the lung capillaries (Fischer et al. 2009). The diameter of MSCs

has also been suggested to function as a physical obstacle for their lung passage

by Schrepfer et al. (Schrepfer et al. 2007). Interestingly, Aicher et al. (2003) reported no lung accumulation of In111-labelled endothelial progenitor cells

(EPCs) when transplanted intravenously or intraventricularly in a rat open-chest

MI model (the physical size of EPCs is considerably smaller in comparison with

the latter two cell types).

In addition to lung entrapment due to the physical size of MSCs and their

relation to the diameter of the pulmonary vasculature, several studies have

suggested involvement of adhesion molecules resulting in attachment of the

transplanted cells to the pulmonary endothelial cells. MSCs are capable of

coordinated rolling behaviour and adhesion onto endothelial cells by binding to P-

selectin and VCAM-1, (Ruster et al. 2006). It has also been suggested that

VCAM-1 is more involved in MSC pulmonary adhesion than P-selectin (Fischer et al. 2009). When comparing the lung clearance of human BM-MSC’s and UCB-

Page 57: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

55

MSC’s in a murine model, the UCB-MSCs had higher expression levels of α4

integrin (CD49d, VLA-4), α6 integrin (CD49f, VLA-6) and the hepatocyte

growth factor receptor (c-Met), as well as a higher general fucosylation level. The

levels of CD49d and CD49f expression could be functionally linked with an

increased lung clearance rate (Nystedt et al. 2013).

Additional evidence supporting the involvement of adhesion molecules in

mediating lung entrapment is that administration of MSCs via two boluses

significantly increased pulmonary passage in a rat model of allogeneous IV

transplantation. The number of MSCs given during the second bolus was

significantly higher as compared to the first bolus, suggesting saturation of

receptors during the first bolus, which allowed more cells to pass through the

vasculature with the second bolus (Fischer et al. 2009). Another factor to be

considered when assessing the relation of physical and adhesion molecule-

mediated lung entrapment of the transplanted cells is that larger cells have larger

cell surface areas with possibly more adhesion molecules expressed per cell,

thereby creating a higher probability for cell-cell contact through adhesion

molecules and their cognate ligands on the alveolar endothelium (Nystedt et al. 2013).

Presumably the lung accumulation of the intravascularly transplanted cells is

a combination of the actual physical entrapment and cell-cell interactions. Passive

circulation of the cells to the lungs can be assumed to lead to the deposition of the

cells within the lungs, but it has to be taken under consideration that MSCs are

shown to actively migrate to injured lung tissue (Lee et al. 2009a, Lee et al. 2009b). As the lungs are a natural barrier with constant exposure to pathogens and

particles of the breathed air, it can be hypothesized that the lungs are under a

constant state of inflammation, thereby facilitating active adhesion of the

transplanted MSCs. What makes understanding the mechanisms and

consequences of the lung entrapment even more complicated are the observed

positive effects after lung entrapment. For example, Lee et al. showed that

hMSCs trapped in mouse lungs attenuated myocardial infarction and the effect

was mediated by TSG-6 (Lee et al. 2009c).

2.5.3 Thromboembolic events

The well documented tendency of the IV transplanted MSCs to accumulate to

lungs in large numbers raises obvious concerns of safety. A number of studies

Page 58: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

56

have confirmed these suspicions, indicating the occurrence of thromboembolic

events after intravascular MSC transplantation.

Thromboembolus formation and MSCs

Blood coagulation results from a cascade that normally begins after the

occurrence of endothelial damage to the vessel wall. Subendothelial layers

contain tissue factor (TF) and certain collagens that trigger the coagulation

cascade. The coagulation cascade consists of primary hemostasis, in which

activated platelets form a clot. Primary hemostasis is followed by secondary

hemostasis, whereupon multiple coagulation factors interact with each other,

forming a cascade that reinforces itself, resulting in the formation of an insoluble

fibrin clot. The secondary hemostasis is initiated by subendothelial collagenases

in the intrinsic pathway or by TF via the extrinsic pathway (van der Windt et al. 2007).

Moll et al. conducted in vitro and in vivo studies investigating interactions

with the coagulation system. MSCs express TF and collagen I, which are both

prothrombotic factors normally found in the subendothelial compartment that

could induce an instant blood-mediated inflammatory reaction (IBMIR) in which

transplanted cells trigger the coagulation cascade. The prothrombotic effects of IV

MSCs in vivo were weak when infused in clinically relevant doses and resulted in

a decrease in thrombocyte counts. The prothrombotic effects increased when late

passage (P5-P8) cells were used (Moll et al. 2012). Intravascularly transplanted

adipose tissue-derived MSCs have been shown to lead to possible formation of

pulmonary emboli triggered by tissue TF (Tatsumi et al. 2013).

Several animal studies have demonstrated adverse thromboembolic events

associated with MSC transplantation. IV transplantation of murine MSCs in a

murine model displayed aggregates of MSCs in the lung as early as one day post-

transplantation, which in many cases persisted thereafter to result in tissue

damage with development of fibrotic tissue, as well as impaired organ function

(Anjos-Afonso et al. 2004). IV transplantation of hMSCs in a murine model led

to lethal pulmonary emboli (Lee et al. 2009d). MSC entrapment at the pre-

capillary level and even microembolus formation has been reported with intra-

arterial transplantation (Toma et al. 2009). Intracoronary injection has also led to

decreased coronary blood flow in porcine models and even myocardial infarction

in canine subjects (Freyman et al. 2006, Vulliet et al. 2004). Clotting and

Page 59: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

57

decreased capillary blood flow is also reported in an intravital microscopy study

(Furlani et al. 2009).

The consequences of MSC-related thromboembolism

Past studies led to an assumption that IV transplanted cells can cause a pulmonary

embolism in a human subject. The consequences of pulmonary embolization vary.

Embolization of pulmonary capillaries decreases the effective ventilation area of

the lungs, leading to hypoxia. Embolization of main pulmonary arteries or a

sufficiently large amount of pulmonary capillaries can lead to failure of the right

ventricle due to increased working load, ultimately resulting in circulatory

collapse collapse (McIntyre & Sasahara 1971).

The effects caused by IV transplanted cells depend on whether they are able

to cause the formation of a large clot, causing embolization of the main

pulmonary artery truncus, or the formation of microemboli in the pulmonary

capillaries. Microembolisation of the capillaries could be tolerated to a certain

extent, depending on the condition of the subject’s heart and ventilation function

collapse (McIntyre & Sasahara 1971). Massive embolization of the main

pulmonary arteries, on the other hand, could cause lethal consequences on an

otherwise healthy subject. Pulmonary embolization that leads to haemodynamic

instability could be treated with anticoagulative agents or thromobolytic therapy

in a normal clinical setting, but these treatment options could be unavailable

under certain clinical conditions with inherent risks of bleeding that MSC therapy

could otherwise cover (e.g. traumatic brain injury).

Thromboembolic events caused by cell transplantation could have other

severe consequences when applied via the IA route. If the cells were targeted to a

certain organ with catheterization techniques and emboli formed emboli (Anjos-

Afonso et al. 2004, Vulliet et al. 2004, Freyman et al. 2006, Furlani et al. 2009

Lee et al. 2009d, Toma et al. 2009), this would naturally result in ischemic

damage and loss of function of the target organ. Again, the type of damage would

depend on whether the cells caused macroclotting or reached the vasculature at

the capillary level. Macroembolus formation after central IA delivery, such as via

the intraventricular route, would resemble the thromboembolic events caused by

atrial fibrillation, in which the embolus is mostly limited to the cerebral

vasculature, ultimately leading to stroke. Other possible consequences of central

macroembolisation would be acute limb ischemia, acute mesentery ischemia or

kidney infarction. If cells were to cause microembolisation after central delivery,

Page 60: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

58

this would presumably lead to sporadic ischemic events throughout the body that

would otherwise hypothetically cause reversible damage, but as the brain is

recognized as the primary target of the central emboli, this could have severe

consequences.

Contrary to the preclinical studies reporting possible associated threats, the

clinical trials with MSCs or BM-MNCs have not revealed the occurrence of

thromboembolic events, but Moniche et al. have reported the occurrence of

seizures during follow-up on two patients who received IA BM-MNC

transplantation for acute stroke, although clear causality could not be indicated

(Moniche et al 2012). Central venous route for BM-MNC administration was

used in a human trial with pediatric patients suffering from traumatic brain injury

(Cox et al. 2011), with no adverse effects. Intravenous transplantation of

autologous MSCs for patients after myocardial infarction also did not reveal

safety threats (Hare et al. 2009). Current clinical studies with intravascular MSC

or BM-MNC transplantation have neglected the lack of sufficient information

regarding the risks of thromboembolic events and have, in fact, endangered the

trial participants, as no salvage plan, in case of occurrence of adverse effects, is

presented. Human MSC transplantation showed no manifestations of acute or

chronic toxicity in adult unconditioned mice in a 13-month follow-up study

(Allers et al. 2004).

2.5.4 Immunologic reactions

Transplantation of allogeneic MSCs could potentially result in adverse

immunologic reactions. The transplantation subject’s immune system could

interpret the transplanted cells as hostile and attack them. As the transplanted cells

are shown to persist in the subject’s tissues for extended periods of time (Allers et al. 2004, Meyerrose et al. 2007, Vilalta et al. 2008), this hypothetically could lead

to the development of a GvHD-type of condition if the transplanted cells targeted

the subject’s tissues. Donor MSC transplantation has been reported to lead to an

inflammatory response in healthy rat graft rejection within 7 days after direct

stereotactic transplantation (Coyne et al. 2006) and dose- and antigenic mismatch

degree-dependent immunogenic reactions were also seen when MHC class I

mismatched MSCs were intracranially transplanted in primates (Isakova et al. 2010). However, when autologous MSCs were transplanted in human subjects

that had developed GvHD after HSC transplantation, only weak immunogenic

potential was observed (Sundin et al. 2009). These immunologic issues can be

Page 61: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

59

avoided by choosing autologous transplantation, but in the case of MSCs, this

would practically rule out the possibility of transplantation in the hyperacute

phase after ischemic injury due to time needed for cell cultivation.

2.5.5 Development of malignancies

Concerns of potential malignant transformation of MSCs have arisen regarding

their transplantation in clinical trials. The MSCs are multipotent progenitors with

clonogenic capabilities and thus possess a theoretical possibility to form

malignant clones. Since the cells are shown to persist in host tissues, an

unavoidable question is whether they can generate malignancies at some point

during their life span. Possible malignant transformation of the engrafted cells

could occur with a delay of decades, which makes it hard to study this potential

threat. Human adipose tissue-derived MSCs in a murine model with an 8 month

follow-up period showed no tumour formation (Vilalta et al. 2008). Long-term

engraftment of MSCs in human subjects appears to be low, hence suggesting the

risk of malignancies to be unlikely (von Bahr et al. 2012). It has been speculated

that autologous cells could have a lower risk of malignant transformation, as they

may be eventually rejected despite their immunoprivileged nature. Currently no

reported malignant transformation of MSCs is known (Barkholt et al. 2013).

In addition to malignant transformation of the transplanted cells, the cells

could accelerate the development of host malignancies (Barkholt et al. 2013). A

common clinical example of this potential cancer cell-MSC interaction is adipose

tissue transplantation for reconstructive breast surgery after mastectomy resulting

from breast cancer. There is evidence supporting the theory that adipose tissue-

derived MSCs can promote the growth of breast cancer cells (Zimmerlin et al. 2011) and their immunosuppressive effects are shown to favour tumour formation

in a murine model (Djouad et al. 2003). It is known that the cells induce

angiogenesis (Chen et al. 2003, Fujita et al. 2010), which is a well-recognized

factor involved in growth of malignant tumours. Immunosuppressive properties

of the MSCs could also interfere with the naïve immune system’s anti-oncogenic

mechanisms.

2.5.6 Ectopic tissue formation

The MSCs are stem cells of mesenchymal origin and a natural concern is that they

are capable of differentiating into various types of mesenchymal tissues. As the

Page 62: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

60

transplanted cells become affected with an uncontrollably varied number of

different cellular signals released by the targeted tissue or the host’s unaffected

system, there is a possibility that the MSCs could differentiate into impractical

tissues, which are unsuitable for the target organ’s function. Indeed, in a murine

model of MI, direct lesional injection of allogeneic MSCs led to the formation of

calcification and ossification, demonstrating a potential safety threat (Breitbach et al. 2007).

The clinical consequences of ectopic tissue formation would depend on the

type of tissue formed and the host organ’s function. Osteogenic differentiation in

myocardial tissue could potentially affect contractile function of the heart and

ectopic formation of neural tissue could hypothetically cause abnormal

conduction circuits within the myocardium, potentially resulting in arrythmias

although no adverse effects in a placebo controlled clinical trial with an

intramyocardial injection was present (Lehtinen et al. 2014). Ectopic tissue

formation within the cerebral parenchyma could potentially lead to seizure

formation. Low long-term engraftment of MSCs in human participants is reported

(von Bahr et al. 2012) and thus it can be considered relatively unlikely that

formation of ectopic, MSC-derived tissue would be a threat, unless this would

happen in critical sites of physiologic function.

2.6 Optimisation of intravascular MSC transplantation

Past research has revealed certain challenges concerning the intravascular

transplantation of MSCs. A range of studies have focused on ways to overcome

these particular issues and improve the efficacy and safety of the transplantation

procedure.

2.6.1 Type of injury

When applying MSCs for ischemic organ injury, the type of injury can be

assumed to remarkably affect the chance of the transplantation’s success.

Individual organs have different microenvironments and secretomes that ischemic

injury greatly affects. Thus, the behaviour of the MSCs should first be studied

with organ-specific preclinical settings and generalized assumptions of MSC

therapeutic functions should be avoided until their exact biological characteristics

are fully understood.

Page 63: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

61

Another possible factor possibly affecting the success of cellular therapy is

the severity of the ischemic injury. Shindo et al. compared transplanted neural

stem cell (NSC) survival between a rat model of mild and severe TBI, concluding

poorer survival with more severe injury (Shindo et al. 2006). This can also be

hypothesized to apply to bone marrow-derived stem cells. It is crucial to explore

the limits of MSCs’ restorative effects, as the therapeutic gain should always

exceed the possible adverse effects. If the severity of the ischemic injury in a

clinical setting prevents the MSCs having any therapeutic benefit, then the use of

cellular therapy should be avoided.

2.6.2 Cell type

The type of the therapeutic cells could have a major impact on the biodistribution

and therapeutic capacity of the cell transplantation. The possibility for rapid re-

transplantation of the BM-MNCs is recognized as a favourable factor for their

use. Again, it is obvious that the cultivated MSC population clearly contains more

actual therapeutic stem cells, leading to a natural presumption that this population

possesses more therapeutic capacity. BM-MNCs also contain HSCs, which have

been suggested to have neuroprotective effects independently due to their

immunomodulatory properties, such as a reduction of immune cell migration to

the ischemic lesion in stroke (Schwarting et al. 2008).

Studies comparing the effectiveness of MSCs and BM-MNCs are rare. The

MSCs and BM-MNCs have been reported to have similar effects when

transplanted early after the onset of focal cortical ischemia (de Vasconcelos Dos

Santos et al. 2010). MSCs have been reported to be more effective in a model of

chronic myocardial infarction (Mazo et al. 2010). The small number of studies

thus favours the use of MSCs for their therapeutic capacity, but again the factors

to be considered are the timing and safety of the transplantation, as the BM-

MNCs could have better pulmonary passage (Schrepfer et al. 2007, Fischer et al. 2009).

The source of MSCs could also affect their biodistribution and therapeutic

capacity. Nystedt et al. compared the characteristics and pulmonary passage of

human umbilical cord MSCs (hUC-MSCs) and BM-MSCs. The hUC-MSCs had

higher expression levels of α4 integrin (CD49d, VLA-4), α6 integrin (CD49f,

VLA-6) and the hepatocyte growth factor receptor (c-Met), as well as a higher

general fucosylation level, and the level of CD49d and CD49f expression could

be functionally linked with increased lung clearance rate (Nystedt et al. 2013).

Page 64: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

62

Although bone marrow acts as an easily accessible source for MSCs, this finding

suggests that MSCs of other sources could possess different biodistribution

capacities.

It is currently not certain whether allogeneic cells have an inferior therapeutic

capacity compared to autologous cells. The allogeneic transplantation could

interact with the host immune system by possibly limiting their biological

function. Antiproliferative effects on lymphocytes have been observed to occur

with both autologous and allogeneic MSCs and involvement of MHCs with

MSCs is speculated to not be required (Le Blanc et al. 2003). This finding

encourages the use of cells of autologous origin as precultivated cells could be

available during the acute phase of the ischemic injury.

2.6.3 Systemic transplantation route

Intravascular transplantation can be performed via IV or IA routes, both of which

could have advantages and disadvantages concerning biodistribution, therapeutic

capacity and also safety. The effectiveness of IV transplantations has been

questioned due to the lung entrapment effect (Fischer et al. 2009).

Although the number of studies comparing these two transplantation routes is

limited, the results generally favour the IA route. IA transplantation resulted in a

better outcome than IV in a rodent model of MCAO (Kamiya et al. 2008). In a

murine model, IA administration of allogeneous adult multipotent progenitor cells

is shown to result in decreased lung accumulation and an increased amount of

cells in the liver, kidneys and brain at 30 days compared with IV transplantation

(Tolar et al. 2006). Choosing to transplant the therapeutic cells by an IA route

could thus enhance their therapeutic effects and biodistribution. Again, the

possible disadvantages of the IA route (i.e. more invasive approach and

embolization), would then be encountered.

2.6.4 Timing of transplantation

The therapeutic window of bone marrow cell transplantation can be hypothesised

to affect cell biodistribution. Using ischemia- reperfusion injury of the brain as an

example, certain factors related with transplantation timing can be recognized.

A rodent study compared the effects of early versus delayed transplantation of

MSC migration in damaged myocardium after intra-arterial transplantation,

showing a statistical trend of higher uptake at 48h after the insult compared to 10-

Page 65: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

63

14 days following transplantation (Barbash et al. 2003). A study utilising a rat

model of multiple focal cortical ischemic lesions demonstrated improvement of

neurologic outcome when BM-MNCs where administered at post-ischemic day

(PID) 1 or 7, but not at PID 14 or 30; the same study also used MSCs with

positive outcome when transplanted at PID 1 but not at PID 30 (de Vasconcelos

Dos Santos et al. 2010). A rat model of MCAO also reported improved recovery

of MSC transplantation at PID 1 and 7 (Chen et al. 2001b). A rat study with IV

transplantation for MCAO presented strong evidence for earlier transplantation

superiority; 3h post-ischemic transplantation had the most therapeutic effect, with

the effect linearly decreasing to 72h (Iihoshi et al. 2004). A rodent cerebral

ischemia model with BM-MNC transplantation investigated the optimal delivery

time, comparing 72h with 7 days administration, and observed neurologic

improvements with earlier transplantation (Yang et al. 2011).

It appears that greater therapeutic effect is achieved if the cells are

administered in the acute phase after the ischemic insult. Favourable results are

generally seen when transplantation is performed within the first 24 to 72 hours

(Barbash et al. 2003, Iihoshi et al. 2004, de Vasconcelos Dos Santos et al. 2010,

Yang et al. 2011), although some benefit may be seen when the cells are

administered within one week after the insult insult (Chen et al. 2001b, de

Vasconcelos Dos Santos et al. 2010). However, the evidence supporting even

earlier transplantation at 3 hours after cerebral ischemia (Iihoshi et al. 2004)

strongly emphasizes the need to consider the use of BM-MNCs due to their

availability without cultivation.

2.6.5 Cell dose

Optimal therapeutic cell dose is not yet fully understood, but there are certain

factors that must be considered. Firstly, the used cell dose has to have clinical

effect. Secondly, the dose should be safe and the consequences of increasing the

dose beyond the minimal effective amount should be carefully considered.

Increasing the dose could result in more therapeutic potential, but this would also

be likely to increase the probability of possible adverse effects related to the

transplantation.

Several studies support the hypothesis that the therapeutic effects have a dose

response. A rat study with MCAO reported improved recovery with high dose of

MSCs for transplantation, but no statistically significant effect was seen with a

lower dose (Chen et al. 2001b). Mouse MSC transplantation in rat MCAO

Page 66: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

64

showed dose-dependent cerebral blood flow (CBF) and blood brain barrier (BBB)

restoration in addition to dose-dependent increases of growth factor secretion;

subjects with cell doses of 2 x105 or 105 cells transplanted directly into the lesion

showed better results than those with cell doses of 104 or 4 x 104 (Borlongan et al. 2004). Wolf et al. conducted a porcine study experimenting with dose ranges of 1

x 103 to 1 x 106 IV transplanted autologous or allogeneic MSCs and demonstrated

a dose-dependent improvement in left ventricular function after MSC delivery,

statistically significant at 1 x 105 and 1 x 106 cells per body weight (kg), as

assessed by echo cardiography (Wolf et al. 2009). A proliferative effect on

lymphocytes is reported in very low MSC doses in vitro, but a dose-dependent

antiproliferative effect was seen with larger MSC quantities (Le Blanc et al. 2003).

On the contrary, some studies have witnessed a lack of dose response or

limitations of dose response by further increasing the dose. Hare et al., conducting a human trial with MI patients receiving IV allogeneic hMSC

transplantation, reported a decrease in ventricular arrhythmias and improved

pulmonary function. Dose response was also evaluated in 3 placebo-controlled

dose escalation cohorts of 0.5, 1.6, and 5.0 × 106 hMSCs/kg body weight, but no

dose-response was observed except for the number of premature ventricular

contractions, which decreased among higher hMSC doses (Hare et al. 2009).

Yang et al. compared 1, 10 or 30 x 106 doses of BM-MNCs transplanted at 24

hours in a rat model of cerebral ischemia and reported no therapeutic benefits

from the 1 x 106 dose. Additionally, increasing the dose up to 30 x 106 from 10 x

106 did not improve the therapeutic benefits (Yang et al. 2011).

Albeit higher cell doses can be assumed to result in better therapeutic

outcome, as the transplanted cells would likely have a higher capacity for

releasing beneficial soluble factors, larger quantities could also have adverse

effects due to the nature of cell biodynamics. Intravascular transplantation of

human MSCs in a mouse model is reported to cause embolus formation in

capillaries and the effect was more prominent with higher cell concentrations

(Furlani et al. 2009). Moll et al. investigated the prothrombotic effects of MSCs

and although the adverse effects were weak when median 1,6 x 106 cells per

kilogram were given at a low cell-passage number (P1-4), they concluded that

higher doses could have more prothrombotic potential (Moll et al. 2012).

Optimal cell dosage is a factor to be determined when considering

intravascular stem cell transplantation. Low dose transplantation has even resulted

in a lack of therapeutic results, although further increasing the number of

Page 67: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

65

transplanted cells in high dose transplantation appears to not offer greater

therapeutic benefit (Yang et al. 2011). It could be speculated that the dose-

response of MSCs could bring greater therapeutic benefit until a certain plateau is

reached. In addition to the lack of a greater benefit gained by further increasing

the dose, the increased risk of thromboembolic events possibly connected with

higher doses becomes a concern. The balance between embolus risk and

biologically potent cell quantity is an important factor that needs to be solved in

future studies.

Another factor to be considered when defining the optimal cell dose is that

fresh bone marrow aspirates of BM-MNCs collected from each individual

contains a varying amount of therapeutic cells and thus it is necessary to

determine whether the amount of re-transplanted cells needs to be manipulated to

optimise the therapeutic outcome. If MSCs are used for treating an ischemic

organ injury, it would be likely that their isolation and cultivation would begin as

the patient would be admitted to the point of care. Recognizing that earlier

transplantation leads to greater therapeutic effects (Barbash et al. 2003, Iihoshi et al. 2004, de Vasconcelos Dos Santos et al. 2010, Yang et al. 2011), it is essential

to specify the cell dose that leads to optimal therapeutic effects, as extending the

cultivation time to produce more cells potentially sacrifices the therapeutic benefit

of earlier transplantation.

Usually, the MSC transplantation is executed using a slow infusion. However,

it is still unclear whether multiple boluses or infusions could have beneficial

results. The administration of 4 x 106 MSCs via two boluses significantly

increased pulmonary passage in a rat model of allogeneous IV transplantation

(Fischer et al. 2009). The result was explained by saturation of lung adhesion by

the first dose. This finding suggests that performing the MSC transplantation

using more than one dose or infusion could offer certain advantages. Qian et al. reported no decrease in pulmonary entrapment with low MSC concentrations

(Qian et al. 2006).

2.6.6 Pronase detachment

Pronase, used as a dispersing agent of cultured cells in the 1970s, is currently

commonly used in protein analysis and antigen retrieval protocols (Foley &

Aftonomos 1970). Pronase has mostly been replaced by trypsin as a dispersing

agent. Given the fact that cell surface adhesion molecules clearly effect lung

deposition, substances that affect cell attachment to surfaces, such as dispersing

Page 68: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

66

agents, could have an impact on cell adhesion characteristics, thereby potentially

affecting lung clearance (Nystedt et al 2013).

2.6.7 Vasodilatators

Due to the fact that pulmonary entrapment of the intravascularly transplanted

therapeutic cells has been correlated to their diameters in relation to lung

capillaries capillaries (Schrepfer et al. 2007, Fischer et al. 2009), vasodilating

agents could thus increase the pulmonary clearance of the transplanted cells by

allowing more cells to pass through the narrow capillaries. Application of sodium

nitroprusside has shown to dilate lung capillaries, thus increasing the lung passage

of MSCs (Gao et al. 2001, Schrepfer et al. 2007). The use of sodium nitroprusside

also increased the MSC accumulation in the liver and long bones as a result of

greater lung clearance (Gao et al. 2001). These are encouraging results, as

vasodilators are commonly used in various clinical settings and their possible

adverse effects, mainly hypotension, is familiar to clinicians, clinically tolerated

and reversible.

2.6.8 Anticoagulative agents

The reports suggesting possible thromboembolic manifestations after MSC

transplantation (Anjos-Afonso et al. 2004, Freyman et al. 2006, Furlani et al. 2009, Lee et al. 2009d, Tatsumi et al. 2013, Toma et al. 2009, Vulliet et al. 2004)

are a safety threat to be considered. Possible pharmaceutical interventions to

prevent thromboembolus formation would primarily consist of using

anticoagulative agents. Tatsumi et al. reported successful suppression of MSC’s

procoagulative properties by applying anti-TF antibody or using factor VII

(Tatsumi et al. 2013). Suspension of IV transplanted MSCs in heparin in a mouse

model was highly beneficial, whereas application of MSCs suspended in

PBS/EDTA or control buffer caused severe pulmonary reactions and, in some

cases, death (Deak et al. 2010).

The use of anticoagulative agents could serve as a promising tool for

reducing the risk of MSC-induced thromboembolic complications. The possible

adverse effects of the thromoboembolic agents could be the risk of bleeding. The

clinical impact related to the increased risk of bleeding would depend on the

clinical setting in which the cells were used; the risk would be well tolerated in

physiological circumstances, but conditions, such as trauma, could alter the

Page 69: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

67

body’s coagulative system’s balance, thus resulting in the restricted use of

anticoagulative substances because of their potential hazards.

2.6.9 Cellular engineering and culture conditions

Altering the characteristic of the transplanted cells prior to the transplantation

could provide a way to improve their biodistribution and therapeutic capacity.

One interesting option to increase the targeting of therapeutic cells would be to

add a specific tag on the cell surface. For example, chemical modification of the

BM-MSC surface with sialyl Lewisx, the moiety for selectin recognition,

improved the homing of MSCs to inflamed tissue (Sarkar et al. 2011). The

homing of MSCs to ischemic myocardium has been successfully increased by

applying a peptide-based tag (Kean et al. 2012), which also improved E-selectin-

mediated adhesion and rolling of MSCs (Cheng et al. 2012). The bone marrow

homing of BM-MSCs was shown to increase with glycan engineering of CD44

(Sackstein et al. 2008).

Altering culture conditions provide another method of manipulating MSC

biodistribution capacity. Low-density culturing increased the expression of

podocalyxin-like protein and CD49f on the cell surface, resulting in better

migration in a MI model (Lee et al. 2009d). Cytokine pretreatment and hypoxic

culture conditions induced migration, engraftment, and therapeutic potential of

MSCs (Hemeda et al. 2010, Hung et al. 2007, Rosova et al. 2008). These

applications could offer potential benefit in the future and enhance the therapeutic

properties and tissue targeting of the MSCs.

Page 70: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

68

Page 71: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

69

3 Aims of the study

I How do BM-MNCs and MSCs distribute to organs when transplanted

intravenously or intra-arterially in an intact porcine model and does the intra-

arterial route decrease lung accumulation? Does intravascular transplantation

of these cells cause pulmonary embolisms?

II Can intra-arterially transplanted BM-MNCs reach the brain after a global

ischemic brain injury and is there a culmination point of migration during the

first 24 hours?

III Does transient proteolytic modification of MSCs with pronase decrease lung

accumulation and improve migration to target organs and how does pronase

affect cell characteristics?

Page 72: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

70

Page 73: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

71

4 Materials and methods

4.1 Experimental animals (Studies I, II, III)

The porcine studies (Studies I-III) were conducted with female domestic pigs

weighing between 18-35 kg. Study III used female Wistar Han rats and 7–8-

week-old female mice. All the studied animals received humane care in

accordance with the “Principles of Laboratory Animal Care” formulated by the

National Society for Medical Research and the “Guide for the Care and Use of

Laboratory Animals,” prepared by the Institute of Laboratory Animal Resources,

National Research Council (published by the National Academy Press, revised in

1996). The porcine studies were approved by the Research Animal Care and Use

Committee of the University of Oulu, Oulu, Finland. The Study III rodent

protocols were approved by the ethical committee of Helsinki University Central

Hospital, the Finnish Red Cross Blood Service and the National Animal

Experiment Board in Finland.

4.2 Perioperative protocol of porcine model (Studies I, II and III)

The animals were sedated with ketamine hydrochloride (15 mg/kg) and

midazolam (2 mg/kg). Anaesthesia was induced with thiopental and fentanyl

followed by endotracheal intubation. The anaesthesia was maintained by a

continuous infusion of fentanyl (25 µg/kg/h), midazolam (0.25 mg/kg/h),

pancuronium (0.2 mg/kg/h), and inhaled isoflurane (1.0%). The animals received

intravenous Ringer’s Acetate (15 mL/kg/h) throughout the experiment.

An arterial catheter was positioned into the right femoral artery for arterial

pressure monitoring and blood sampling. A thermodilution catheter (CritiCath, 7

French [Ohmeda GmbH & Co, Erlangen, Germany]) was installed via the right

femoral vein into the pulmonary artery, allowing sampling of blood, monitoring

of central venous pressure, pulmonary artery pressure and pulmonary capillary

wedge pressure, and the recording of blood temperature and cardiac output. Mean

arterial pressure was maintained at a minimum level of 65 mmHg with fluids and

norepinephrine infusion.

Arterial blood gases, pH, electrolytes, serum ionised calcium, glucose,

haemoglobin levels (i-STAT Analyzer, i-STAT Corporation, East Windsor, NJ),

basic blood count, leukocyte differential count (Cell-Dyn Analyzer [Abbot, Santa

Page 74: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

72

Clara, CA]), creatine kinase-MB ([Hydrasys LC-electrophoresis, Hyrys-

densitometry, Sebia, France]) and troponin-I (Immunoassay, Innotrac AIO, Turku,

Finland) were measured at the baseline.

4.3 Porcine MSCs and BM-MNCs (Studies I, II and III)

Animals assigned to receive BM-MNC’s underwent bone marrow harvesting after

the induction of anaesthesia resulting in autologous transplantation. Bone marrow

was harvested from both tibial bones with a sternal puncture needle and the

aspirate was collected into syringes containing 10 000 IU’s of heparin sulphate.

The bone marrow aspirate (1:5 dilution in phosphate-buffered saline [PBS],

Sigma-Aldrich) was subjected to density gradient centrifugation (Ficoll-Paque

Plus, Amersham Biosciences) to exclude erythrocytes and granulocytes. Bone

marrow mononuclear cells were collected from the interphase and washed 3 times

with PBS. The cells were suspended in a saline solution at a density of 25-50 x

106 cells/mL. Trypan blue dye, which was added to a fraction of isolated cells,

confirmed viability of more than 99% of the transplanted cells.

Bone marrow aspirate was collected and MNCs were isolated from a single

healthy donor animal, as described in the previous section. BM-MNCs were

plated in a cell culture flask at a density of 300 000 cells per cm2 in Minimum

Essential Medium Eagle alpha modification (Sigma-Aldrich) supplemented with

10% fetal calf serum (Promo Cell), 100 U/ml penicillin, 0,1 g/l streptomycin, 20

mM HEPES and 2 mM L-glutamine (all reagents from Sigma-Aldrich unless

otherwise indicated). The cells were allowed to attach for two days after which

the medium was replaced. Half of the medium was replaced every 3-4 days and

the cells were passaged at near confluence. The cells were used for

transplantation at passages 3 or 4.

4.4 Human MSCs (Study III)

Umbilical cord blood (UCB) collections were performed at the Helsinki

University Central Hospital (HUCH) and Helsinki Maternity Hospital (Finland).

Bone marrow (BM) was collected from an unaffected bone site from patients

operated on for osteoarthritis (> 50 years of age) or scoliosis (< 20 years of age).

All donors gave their informed consent prior to sample collection, and the study

protocols were approved by the ethical committee of HUCH, the Finnish Red

Cross Blood Service (FRCBS), and Oulu University Hospital (Finland). Human

Page 75: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

73

UCB-MSC cultivation was performed, as previously described (Laitinen et al. 2011). BM-MSC cultivation was performed, as described previously (Leskela et al. 2003).

4.5 Rat MSCs (Study III)

Wistar rat MSCs (Cyagen Biosciences Inc., Sunnyvale, CA, http://www.cyagen.

com) were cultured according to the manufacturer’s instructions.

4.6 Pronase detachment and cell viability (Study III)

Pronase is a nonspecific protease mixture isolated from Streptomyces griseus. The

subconfluent cells were detached with 0.5% pronase (Roche, Mannheim,

Germany, http://www.roche.com) in phosphate-buffered saline/0.25 mM EDTA.

Different concentrations of pronase, ranging from 0.05 to 1%, were also tested.

Trypsin (TryPLe Express; Life Technologies, Paisley, U.K., http://www.lifetech.

com) detachment was always used as a control. As with trypsin detachment,

pronase detachment was always stopped with excess culture medium within 4

minutes from the time of the addition of the enzyme to protect the detached cells.

Subsequent centrifugation and cell re-suspension were performed to remove the

enzymes. Cell viability was determined by trypan blue exclusion or analysis using

a Nucleocounter NC-100 (Chemometec, Lillerod, Denmark, http://www.

chemometec.com).

Viability was routinely determined in 30 individual pronase-detached cell

samples. Cell morphology was observed and documented by microscopy. For the

protein recovery studies, cells were incubated in culture medium for 5–7 hours at

37°C after detachment with subsequent cell viability measurements. Gentle

agitation of cells was done to prevent the attachment of cells. In addition, the

depolarization of mitochondrial inner membrane potential (ΔΨm) was analyzed

from differently detached BM-MSCs. Re-adherence to culture vessels and growth

kinetics of pronase-detached cells were also investigated and compared with

trypsinized cells.

4.7 Cell surface analysis by mass spectrometry (Study III)

UCB-MSCs were used for mass spectrometry (MS) analysis of cell surface

proteins after trypsin and pronase detachment and processed directly or after 5

Page 76: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

74

hours of post-detachment recovery. The cell surface proteins were biotinylated.

The labelled proteins were harvested with magnetic beads prior to liquid

chromatography (LC)-MS analysis.

4.8 Cell surface analysis by flow cytometry (Study III)

UCB-MSCs and BM-MSCs were analysed for cell surface epitope expression

immediately after use of different detachment protocols and after 5–7-hours of

post-detachment recovery. The antibodies against the following proteins were

used: CD44, CD49d, CD49e, CD73, CD90, CD105, HLA-DR, CD14, CD19,

CD34, CD45, CD13, CD29, CD49c, CD54, CD59, CD106, CD146, CD147

(Abcam, Cambridge, U.K., http://www.abcam.com), CD166, CD184, fibronectin

(FN) (Abcam), galectin-1 (Acris Antibodies, Herford, Germany, http://www.acris-

antibodies.com), and chondroitin sulfate proteoglycan (CSPG)-4. All antibodies

were purchased from BD Biosciences (San Diego, CA, http://www.bdbiosciences.

com) unless stated otherwise.

The cells were labelled with 2 µl or 0.5–1 µg (CD147, FN, galectin-1,

CSPG4) of the antibodies per 1 x 105 cells. Secondary antibody staining was

performed for CD147, FN, galectin-1, and CSPG4. The labeled cells were run

with a FACS Aria (BD Biosciences) flow cytometer, and the results were

analysed with FACSDiva Software (BD Biosciences). Appropriate isotype control

antibodies were also used.

4.9 T-cell proliferation assay (Study III)

Peripheral blood mononuclear cells (PBMNCs) were isolated from buffy coats

from anonymous blood donors (FRCBS) by density gradient centrifugation

(Ficoll-Paque Plus, GE Healthcare, Piscataway, USA) and labeled with 5 µM

5(6)-carboxyfluorescein diacetate N-succinimidyl ester (CFSE) solution

(Molecular2 Pronase Detachment of MSCs Probes, Eugene, OR,

http://probes.invitrogen.com). CFSE-labeled PBMNCs were cocultured with

trypsin- or pronase-detached UCB-MSCs (n = 2) in RPMI with 10% fetal bovine

serum (FBS), 100 U/ml penicillin + 0.1 mg/ml streptomycin (Life Technologies).

MSC:MNC ratios of 1:10, 1:20, 1:50, and 1:100 were used. To activate T-cell

proliferation, 100 ng/ml of the anti-human CD3 antibody clone Hit3a

(BioLegend, San Diego, CA, http://www.biolegend.com) was used. Non-

stimulated CFSE-labelled cells and stimulated/nonstimulated non-labelled

Page 77: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

75

PBMNCs were used as controls. T-cell proliferation was analysed after 4 days as

dilution of CFSE intensity by FACSAria flow cytometry (BD Biosciences) and

FlowJo software (version 7.6.1; Tree Star, Ashland, OR, http://www.treestar.com).

4.10 Angiogenesis assay (Study III)

Angiogenesis was studied in a validated angiogenesis co-culture model (Sarkanen et al. 2011) using human fibroblasts (American Type Culture Collection, Boras,

Sweden, http://www.atcc.org) and human umbilical cord vein endothelial cells

(HUVECs). Fibroblasts and HUVECs were co-cultured for 7 days before adding

differently detached UCB-MSCs. UCB-MSCs were also plated in inserts

(Scaffdex, Tampere, Finland, http://www.scaffdex.com). The following media

were used: basic test medium (BTM) (Endothelial Basal Medium-2 with 2.0%

FBS, 0.1% Gentamicin, Amphotericin-B, 1% L-glutamine; Lonza Group Ltd.,

Basel, Switzerland, http://www.lonza.com), BTM with angiogenic growth factors

for positive controls, and UCB-MSC medium. Technical validity was ensured

with positive (n= 4) and negative controls (BTM, n= 3; UCB-MSC medium, n=

2). On day 13, tubules were visualized with anti-von Willebrand factor staining

and analysed as described (Sarkanen et al. 2011).

4.11 In vitro rolling assay (Study III)

A Diaphot TMD inverted-stage microscope (Nikon, Tokyo, Japan,

http://www.nikon.com) configured with phase contrast, objective lens (4x),

MicroPublisher 5 megapixel camera (QImaging, Surrey, BC, Canada, http://www.

qimaging.com) and MCID Core image analysis program (InterFocus Imaging

Ltd., Cambridge, U.K., http://www.mcid.co.uk) was used to count the attached

cells and their average rolling speed on FN-coated (0.5 µg/µl) capillaries

(diameter, 0.93 mm; length, 12.5 cm). An Alaris-Asena syringe pump

(CareFusion Corp., Palm Springs, CA, http://www.carefusion.com) was used for

perfusion with a calibrated flow speed (5 ml/hour). The cells (20,000 cells per

milliliter) were imaged for 2 minutes in each analysis.

4.12 Labelling of the porcine cells (Studies I, II and III)

The cells were labelled with Tc99m-HMPAO (Ceretec®, Amersham Healthcare)

using a standard technique for leukocyte labelling (Vorne et al. 1989). In brief,

Page 78: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

76

534 – 702 MBq 99mTc-HMPAO was added to the BM-MNC suspension, and left

for 15 – 20 min at room temperature. The cell suspension was centrifuged in

sterile tubes at 300 x G for 10 min. The supernatant was then separated from the

BM-MNCs, and the cells were re-suspended in 2 ml saline.

The whole BM-MNC population was first labelled, then the dose

standardised to the weight of the animal (5 x 106 cells/kg) was isolated and

suspended in 10 cc of saline. In the subjects with MSC transplantation, the

amount of cells that first underwent labelling was 3-6 x106 and the standardised

dose was 5 x 105 cells/kg. Control samples of 50 000 and 100 000 were

administered into vials for later use as gamma counter references. After the

standardised dose was rationed, the activity of the syringe was measured. An

activity level of 90 MBq was set as a requirement, as lower activity could

compromise radionuclide imaging quality. If the threshold activity level was not

reached, the experiment was terminated.

4.13 Intravenous transplantation (Studies I, II and III)

An intravenous route was established by installing a Schwan-Ganz-type

triluminal pulmonary arterial catheter. An incision was made in the left groin in (I,

III) or the right groin (II). Femoral artery and vein were exposed and ligated. The

catheter was carefully inserted from an incision to the vessel wall. Location of the

tip of the catheter was confirmed by typical pressure curve and wedge curve

caused by an inflated balloon (this described method is commonly used in clinical

practice). The cells were administered by applying the lumen with an opening

positioned in the right atrium. After slow injection, the catheter was then rinsed

with 50 cc of saline.

4.14 Intraventricular transplantation (Study I)

An intraventricular route was established by installing a coronary arterial catheter

into the left ventricle. The catheter was inserted through the left femoral artery

after open surgical preparation. Arterial pressure was measured in these animals

using the catheter instead of an arterial needle. Fluoroscopic imaging and typical

pressure curve were used to confirm the intraventricular location. The cells were

transplanted through the arterial catheter followed by rinsing of the catheter with

saline. After the transplantation, the tip of the catheter was then retracted from the

ventricle to avoid prolonged interference of aortic valve function.

Page 79: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

77

4.15 Ischemic brain insult (Study II)

A left thoracotomy was performed from the second intercostal space. The

brachiocephalic trunk and the left subclavian artery were clamped for a period of

7 minutes after systemic application of heparin. The response to sustained

cerebral blood flow was assessed from the rapid depression of BSR in EEG,

which was considered as validation of significant brain ischemia. After releasing

the clamps, 120 mg papaverinehydrophosphate was applied to the vessels.

4.16 Radionuclide imaging (Studies I, II and III)

The animals were kept under anaesthesia for 8 hours followed by transfer for

radionuclide imaging. Hemodynamic parameters and vital functions were

monitored and maintained during the follow-up period. Radionuclide images were

acquired using a hybrid camera combining a gamma camera with a CT camera

within the same cantry (Symbia T2; Siemens Medical Solutions USA, Inc.). A

planar whole-body scan was performed after the follow-up period, with both

anterior and posterior images being acquired at a speed of 5 cm/min using low-

energy high-resolution collimators, a 256 × 1024 matrix, and a 140 keV ± 20%

photopeak (I, II and III). After a planar scintigraphy, the SPECT study was

obtained over a 360º arc, using 96 frames at 90 s per frame. The images were

acquired into a 128 × 128 matrix. The CT part was acquired at a slice step of 3

mm, a current of 120 mAs, and a voltage of 130 kV. Whole body SPECT (I, III)

or head SPECT only were acquired (II).

4.17 Image and data analysis (Studies I, II and III)

Dual-head whole-body acquisitions were used to calculate geometric mean

(Gmean) datasets to estimate organ uptake of BM-MNCs (I, II) and MSCs (I, III).

Regions of interest (ROI) were drawn around the lungs, liver, spleen, kidneys,

and whole body. Gmean was calculated for the different ROIs using the formula

Gmean = square root (anterior × posterior), in which the anterior is the number of

counts from the anterior projection and the posterior is the number of counts from

the posterior projection. Mean activity of individual organ ROIs were normalised

to whole-body mean activity to achieve comparable data between subjects.

SPECT images were analysed by Hermes Hybrid PDR software (Hermes

Medical Solutions, Stockholm, Sweden). ROIs were drawn around target organs

Page 80: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

78

according to anatomic location, as assessed by CT. The ROIs were then copied to

the SPECT data, so their positions and areas were equal. Target organ ROIs were

drawn on preset single slices with an aim to minimize variation between subjects;

the slices were chosen based on clear recognition for repeatability and maximal

avoidance of scattering activity of nearby organs. Mean activity values of ROIs,

provided by analysis software, were used in data analysis to compare individual

organs.

When assessing the lung accumulation (I, III), the ROIs were drawn in all

slices presenting lung tissue. The ROIs were then combined to form a volume-of-

interest (VOI), which was copied to SPECT data, and VOI’s mean activity was

calculated using VOI’s total activity and cell volume. Formation of atelectasis

was confirmed by a radiologist’s expert opinion and VOI of atelectasis areas were

individually measured. To assess the atelectasis’ impact on lung accumulation,

lung VOI mean activity was also calculated after reducing the atelectasis’ VOI

total activity and volume from the corresponding whole lung values, resulting in

mean activity of non-atelectatic lung parenchyma.

4.18 Pulmonary embolism CT- scan (Study I)

In addition to the twelve animals forming the primary study groups, a total of four

pigs underwent a lung embolism CT-scan with radiocontrasting agent infusion

after performing the radionuclide imaging and preceding study protocol. Two (2)

of the animals received BM-MSC transplantation intravenously and two (2)

underwent sham operation with cell transplantation replaced with a similar

volume of saline infusion. The animals were euthanized after the CT-scan and no

tissue biopsies were collected because the execution of the CT-scan could

compromise the acquired data due to infusion of radiocontrasting agent with

possible adverse effects on kidneys that could bias the cell distribution.

4.19 Tissue biopsies (Studies I, II and III)

The animals were euthanized by intravenous injection of pento-barbital. An

autopsy was performed and biopsies were acquired from target organs. Biopsies

were collected from both cerebral hemispheres, the cerebellum, the thymoid

gland, the right atrium of the heart, the apex of the heart, the liver (2 biopsies), the

spleen (2 biopsies), the mesenteric lymph nodes and both kidneys. The biopsies

were inserted into vials and placed in an automated gamma counter (Wallac

Page 81: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

79

Wizard 1480, Perkin Elmer, Gaithersburg, MD, USA). Counts were also

measured from the control samples containing 50 000 and 100 000 cells and used

to calculate biopsy activity index to compare subjects based on biopsy sample

weight and measured activity of control samples and the biopsy sample (results

expressed as counts per minute) (CPM) (I, III).

The following formula was used:

Activity index = (organ CPM / organ weight) / ((50 000 cells CPM / 50 000) + (100 000 cells CPM / 100 000) / 2) = cell count / weight (g) of target organ.

4.20 Electroencephalography (Study II)

Electroencephalography (EEG) electrodes were inserted into the skull. Four

electrodes consisted of two recording electrodes, a reference electrode and a

grounding electrode. Recording began 5 minutes before the ischemic insult and

this period was used as a baseline reference of the burst-suppression ratio (BSR).

Data from EEG were constantly recorded with a NeuroScanSynamps Amplifier,

equipped with NeuroScan 4.0 Software (Neuro Soft Inc., Sterling,VA), using a

sampling rate of 5000 Hz. Isoflurane anaesthesia was maintained constant at 1%

to prevent alteration in BSR. Recovery was assessed by the time it took the

subjects to reach 50 and 100% of the baseline BSR.

4.21 Biodistribution of human MSCs in a mouse model (Study III)

Differently detached UCB-MSCs (p4) and BM-MSCs (p6) werelabelled with

Tc99m-HMPAO (Ceretec; GE Healthcare, http://www.gehealthcare.com) for 15

minutes at room temperature followed by determination of cell viability.

Subsequently, 7–8 week-old female mice received 5 x 105 cells IV in 100 µl of

saline (UCB-MSCs: n= 3; BM-MSCs: n= 5). One hour or 15 hours post-injection

mice were sacrificed, and lungs, heart, liver, spleen, pancreas, kidneys,

gastrointestinal (GI) tract, and femur were collected. Radioactivity of the tissues

was determined with a gamma counter (Wallac Wizard 1480; PerkinElmer,

Gaithersburg, MD, http://www.perkinelmer.com). The relative radioactivity for

each tissue was calculated as a percentage from the total amount of radioactivity

in all the tissues measured. Additionally, the biodistribution of UCB-MSCs

labelled with the fluorescent dye PKH-26 was studied in mice.

Page 82: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

80

4.22 Biodistribution of allogeneic BM-MSCs in a rat model of inflammation (Study III)

Female Wistar Han rats were used in the study. λ-Carrageenan (Sigma-Aldrich,

St. Louis, MO, http://www.sigmaaldrich.com) was dissolved in saline to give a

4% solution, and 150 µl was administered into the plantar region of the left hind

paw immediately before cell injection. Subsequently, animals received either

trypsin-detached (n= 3) or pronase-detached (n=4), Tc99m-HMPAO-labelled rat

BM-MSCs (1 x 106) into the tail vein. Whole-body SPECT-CT (nanoSPECT/CT;

Bioscan Inc., Washington, DC, http://www.bioscan.com) images were acquired

50 minutes, 3 hours, 5 hours, and 24 hours post-injection.

Only one animal per group was imaged at the 24-hour time point, and it was

excluded from the statistical analysis. The volume of the inflamed paw was

measured with a plethysmometer (Ugo Basile, Comerio, Italy, http://

www.ugobasile.com) immediately before and 1 minute after the injection and

directly after imaging. SPECT images were reconstructed with HiSPECT NG

software (Scivis GmbH, Gottingen, Germany, http://www.scivis.de) and fused

with CT data sets using InVivoScope software (Bioscan). VOIs were drawn

around lungs, kidneys, paw, and liver. The same VOIs were used for all

subsequent analyses of corresponding organs. The radioactivity in each VOI was

calculated and corrected for decay. Results are presented as a percentage of the

injected dose.

4.23 Statistical analysis (Study I, II, and III)

IBM SPSS Statistics version 20.0 was used for statistical analysis of the data. The

study groups were compared on transplantation route and cell lineage as 6 versus

6- setup with nonparametric Mann- Whitney U-test for calculating p-values.

Statistically significant differences were considered at p < 0,05. Medians with a

25-75 percent interquartile range were used when reporting values with 6 versus

6- setup. Further comparison was made between subgroups of the same cell

lineage or transplantation route with 3 versus 3- setup; value ranges were reported

and used to assess differences between the subgroups. Data for Study III were

processed with Mascot Distiller (version 2.3; Matrix Science Ltd., Boston, MA,

http://www.matrixscience.com) and searched with Mascot Server (version 2.2.06;

Matrix Science) against human proteins in the UniProtKB database (version

15.12). LC-MS differential expression analysis was performed with Progenesis

Page 83: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

81

LC-MS software (version 2.6; Nonlinear Dynamics Ltd., Newcastle upon Tyne,

U.K.).

Page 84: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

82

Page 85: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

83

5 Results

5.1 IA versus IV transplantation of BM-MNCs and MSCs (Study I)

There were only small differences in the biodistribution patterns between the

three nuclear medicine methods (Tables 5-7, Figures 2 and 3). The effect of the

administration route was smaller in the BM-MNC lineage than in the BM-MSC

lineage. The most striking difference between the two lineages was the

accumulation of the cells in the spleen and vertebrae, with the MNCs having a

much higher accumulation in those organs. The BM-MSCs, in turn, had much

higher uptake in the kidneys. As expected, the majority of the IV-transplanted

BM-MNCs localised to the lungs, followed by the liver, spleen and kidneys. The

IV administration of BM-MSCs resulted in the highest deposition in the lungs,

followed by kidneys, liver, with an equal accumulation being found in the spleen

and vertebrae. The IA route decreased BM-MSC uptake in the lungs (Figure 2)

and increased it in other organs, especially the liver. The IA administration route

did not have such a major impact on BM-MNC lung deposition and its effect on

the other organs was variable. Interestingly, BM-MNC and BM-MSC uptakes

were much higher in the right lung than in the left lung. The atelectic areas had an

approximately two-fold deposition of BM-MNCs and BM-MSCs compared to the

non-atelectic areas. There was no evidence of between-group differences in the

number of atelectic areas.

5.1.1 Computed tomography

Those animals, which underwent helical contrast-enhanced CT did not show any

signs of pulmonary embolism, but they had some atelectic lung areas, as

confirmed by two radiologists.

Page 86: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

84

Table 6. Biodistribution pattern, as assessed from the planar whole-body images.

Lineage Route Lungs Liver Spleen Kidneys

BM-MNCs IV 24-30 4-9 3-7 1-2

IA 16-21 11-14 5-7 1-1

BM-MSCs IV 27-31 2-3 0-2 3-4

IA 13-17 9-12 1-2 6-7

Geometric means were used to assess percentage organ activity of the whole body geometric mean. BM-

MNCs: bone marrow-derived mononuclear cells; BM-MSCs: bone marrow-derived mesenchymal stem cells; IV:

intravenous; IA: intra-arterial (n=3 in each group).

Table 7. Biodistribution pattern, as assessed from SPECT/CT images (percentage of

input activity).

Lineage Route Lungs Liver Spleen Kidneys Vertebrae

BM-MNCs IV 4.7-11.3 1.6-2.8 2.9-4.8 0.4-1.5 2.2-3.5

IA 3.6-6.4 2.5-4.1 2.8-4.7 0.6-1.0 1.2-2.5

BM-MSCs IV 3.6-7.0 0.3-0.5 0.1-0.1 1.1-3.2 0.1-0.1

IA 2.3-3.2 1.3-2.0 0.3-1.0 1.8-4.2 0.2-0.4

BM-MNCs: bone marrow-derived mononuclear cells; BM-MSCs: bone marrow-derived mesenchymal stem

cells; IV: intravenous; IA: intra-arterial (n=3 in each group).

Table 8. Biodistribution pattern, as assessed from activity in tissue biopsies (activity

index).

Lineage Route Right lung Left lung Liver Spleen Kidneys

BM-MNCs IV 7296-11840 3675-7835 1586-2158 3556-6161 731-1119

IA 3138-6618 2446-6579 2620-3418 3885-6363 1051-1618

BM-MSCs IV 8348-10450 2497-11010 209-348 63-77 2709-3689

IA 3118-4943 1155-2323 1246-1759 515-1807 3209-4622

BM-MNCs: bone marrow-derived mononuclear cells; BM-MSCs: bone marrow-derived mesenchymal stem

cells; IV: intravenous; IA: intra-arterial (n=3 in each group)

Page 87: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

85

Fig. 2. Posterior whole body gamma camera images acquired after intravenous (IV) or

stromal cells (BM-MSC) or bone marrow mononuclear cells (BM-MNC).

Page 88: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

86

Fig. 3. SPECT- CT analysis of accumulated cells in the lungs and bone at 8 h. Range

bars in A demonstrate the range of lung accumulation. In panel B, a similar analysis

was performed for the L3 and L5 vertebrae.

Page 89: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

87

Fig. 4. Summary of biodistribution data from planar scan images, SPECT/CT, and

biopsy gamma counter analysis. The biodistribution of transplanted cell at 8 h was

analyzed in all major organs. Range bars represent individual organ values for each

subgroup.

5.1.2 Haemodynamic data

Comparison of subgroup baseline characteristics is presented in Table 8, with no

major differences being observed. None of the study animals showed evidence of

pulmonary hypertension in combination with other symptoms of embolus

formation; thus it was concluded that none of the animals developed

haemodynamically significant pulmonary emboli. Hypotension was observed in

some of the animals after prolonged anaesthesia and blood pressure was

maintained constant with crystalloid infusion and L-norepinephrine and dopamine

infusion if needed. Inotropic support was given to 2 of the animals in the IV BM-

MNC group, 2 of the animals in the IA BM-MNC group and 1 of the animals in

the IV BM-MSC group. The underlying cause of hypotension was clinically

diagnosed to result from possible infection or reaction to the used anaesthetic

agents, and therefore did not appear to be related to cell administration in any of

the animals.

Page 90: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

88

Table 9. Baseline characteristics of study groups (Study I).

Route IV IA

Lineage BM-MNCs BM-MSCs BM-MNCs BM-MSCs

Weight 22.3-25.3 20.6-25.6 20.5-23.8 18.1-23.3

Heart rate 106-117 73-132 96-139 89-150

RRsys 85-101 101-151 69-133 100-140

RRdia 37-54 52-95 57-85 62-77

MAP 54-71 71-118 63-108 78-106

PAsys 14-19 19-29 27-57 20-22

PAdia 7-14 11-22 21-44 10-14

CVP 3-7 4-9 8-9 5-10

PCWP 3-4 1-9 12-14 3-8

C.O. 2.66-3.16 2.47-6.50 2.19-3.97 2.75-3.52

TPA 38.8-39.7 38.4-40.5 36.6-38.5 38.1-38.9

pH 7.52-7.55 7.42-7.53 7.35-7.55 7.52-7.85

pCO2 5.02-5.89 4.90-7.21 5.130-6.43 3.130-5.08

pO2 29.7-34.0 24.6-29.6 32.5-42.0 22.4-38.1

Weight (kg), Heart rate (beats per minute), RRsys = systolic blood pressure (mmHg), RRdia = diastolic blood

pressure (mmHg), MAP = mean arterial pressure (mmHg), PAsys = pulmonary artery systolic blood

pressure (mmHg), PAdia = pulmonary artery diastolic blood pressure (mmHg), CVP = central venous

pressure (mmHg), PCWP = pulmonary capillary wedge pressure (mmHg), C.O. = cardiac output, TPA =

pulmonary arterial temperature (C°), pCO2 = arterial blood carbon dioxide partial pressure (kPa), pO2 =

arterial blood oxygen partial pressure (kPa).

5.2 BM-MNC transplantation for global ischemic brain injury during

first 24 hours (Study II)

Severe but non-lethal ischemia was chosen based on 4 animals that were used as

pilots; clamp intervals tested were between 5 and 15 minutes. Rapid burst

cessation was observed after clamping in all animals. Median time to reach

complete EEG silence was 0:00:20 (0:00:18-0:00:23). The BSR remained at zero

level in all animals after the insult, slowly recovering to 50% at a median time of

0:30:00 (0:25:00-0:45:00) and to a level of 100% at a median time of 1:45:00

(1:25:00-2:00:00). Two animals with a noticeably faster BSR silencing rate also

failed to recover BSR to baseline levels. Recovery data was lost from one subject

due to data corruption.

Page 91: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

89

5.2.1 Proportional counts from biopsies

Activity was divided by biopsy weight, and results were used to form an activity

index for the purpose of easing interpretation and comparability between subjects.

Mesenteric lymph node biopsies had minimal variation between subjects and

were thus chosen as a reference. The activity index was calculated by dividing the

target biopsy index by the mesenteric index and then multiplying by one hundred.

Brain activity proportion remained clearly below 0.6 % for all organs in all

animals. There was no trend towards a higher level of migration nor were major

differences in biodistribution observed in any of the follow-up groups at any

given time. At 4 and 6 hours, however, the number of cells was the highest. The

most up-take was observed in the lungs, the liver and the spleen (Table 9, Figure

5). No visual sign of migration to brain tissue could be detected from either planar

scintigraphy or SPECT-CT reconstructions of the head using the previously

described imaging equipment and image reconstruction software (Figure 6).

Activity could be observed in the skull bones and vertebrae, but no sign of the

transplanted cells could be detected within the brain parenchyma in any of the

subjects. Organ Gmean values were compared with the whole body values (Table

10).

Page 92: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

90

Ta

ble

10

. N

orm

ali

sed

bio

ps

y c

ou

nts

Follo

w-u

p R

ight

cer

ebru

mLe

ft ce

rebr

um

Cer

ebel

lum

R

ight

lung

Le

ft lu

ng

Thym

oid

Live

r S

plee

n

2h

4.1

0.1%

3,

6 0.

1%

3,9

0.1%

12

6036

%

332

10%

62

1.

8%

552

16%

12

70

36%

2h

7.4

0.1%

5,

1 0.

1%

6,0

0.1%

19

9018

%

3940

35

%

188

1.7%

14

2013

%

3730

33

%

4h

10.7

0.

2%

13,3

0.2%

33

,80.

6%

1820

30%

14

80

24%

78

1.

3%

1010

17%

16

50

27%

4h

13.8

0.

1%

15,5

0.1%

18

,20.

1%

5120

31%

74

40

45%

56

0.

3%

1240

8%

2790

17

%

6h

51.6

0.

2%

38,4

0.2%

26

,60.

1%

8590

33%

10

600

41%

46

0.

2%

1530

6%

4980

19

%

6h

37.5

0.

3%

17,6

0.1%

7,

6 0.

1%

3730

29%

42

30

33%

28

0.

2%

658

5%

4318

.133

%

12h

7.4

0.1%

6,

4 0.

1%

8,4

0.2%

13

0023

%

2240

40

%

19

0.3%

39

6 7%

16

80

30%

12h

10.7

0.

1%

10,1

0.1%

13

,10.

1%

1100

10%

55

20

52%

53

0.

5%

1350

13%

25

90

24%

Page 93: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

91

Follo

w-u

p R

ight

cer

ebru

mLe

ft ce

rebr

um

Cer

ebel

lum

R

ight

lung

Le

ft lu

ng

Thym

oid

Live

r S

plee

n

24h

20.7

0.

1%

31.7

0.2%

30

.20.

1%

7470

34%

79

10

36%

43

0.

2%

1780

8%

4500

21

%

24h

11.1

0.

1%

9.0

0.1%

13

.70.

1%

3410

31%

36

80

33%

12

8 1.

2%

367

3%

3500

32

%

Mea

n 10

.9

0.1%

11

.70.

1%

13.4

0.2%

27

0028

%

4090

35

%

54

0.8%

11

2010

%

3150

27

%

Row

s re

pres

ent e

ach

of th

e ex

perim

enta

l sub

ject

s. E

ach

subj

ect's

org

an a

ctiv

ity in

dexe

s ar

e di

spla

yed

in th

e le

ft co

lum

ns. P

erce

ntag

es o

f eac

h ac

tivity

inde

x

com

pare

d to

the

sum

of e

ach

subj

ect’s

inde

xes

are

disp

laye

d on

the

right

. col

umns

Page 94: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

92

Fig. 5. Box-plots demonstrate each organ’s percentage of the whole body value

calculated from the radionuclide planar image in Study II

Fig. 6. Head SPECT-CT acquired from a subject with cell transplantation at 24h time

point shows no sign of cell accumulation within the brain parenchyma in Study II. The

image is scaled to it’s individual maximum, which results cell uptake to the vertebrae

appear prominent.

Page 95: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

93

Table 11. Percentages by organs of the whole body activity

Follow-up Brain Lungs Liver Spleen Kidneys

2h 0% 43.5% 14.3% 2.2% 4.2%

2h 0% 31.0% 16.4% 11.3% 1.7%

4h 0% 34.7% 16.9% 1.8% 2.7%

4h 0% 28.2% 12.1% 8.4% 3.2%

6h 0% 53.4% 12.6% 3.6% 1.2%

6h 0% 33.1% 14.2% 9.4% 2.1%

12h 0% 29.5% 11.8% 11.4% 3.6%

12h 0% 32.3% 17.9% 4.9% 2.3%

24h 0% 36.5% 20.9% 6.2% 2.8%

24h 0% 31.9% 9.0% 11.2% 2.0%

Mean 0% 32.7% 14.2% 7.3% 2.5%

5.2.2 Haemodynamic and biochemical data

The mean arterial pressure, central venous pressure, cardiac output and

pulmonary capillary wedge pressure in all follow-up groups remained comparable

throughout the experiment. No significant differences were detected between

study groups in leukocyte counts, haemoglobin, glucose, serum ionised calcium,

sodium, potassium or cardiac enzymes.

Page 96: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

94

5.3 Effects of MSC pronase detachment on lung clearance and organ targeting (Study III)

The pronase detachment protocol produced viable, single-cell MSC suspensions

without any cell aggregates with > 90% viability (n > 30), which is equal to the

trypsin control. All tested pronase concentrations (0.05–1%) detached the cells

effectively within 4 minutes. The highest concentration (1 %) affected the cell

morphology slightly, although cell viability remained at > 90%. A concentration

of 0.5% pronase was chosen as a standard concentration, as it detached cells

rapidly with high viability and effectively cleaved certain cell-surface proteins.

Pronase-detached cells also adhered normally to culture vessels, proliferated, and

reached confluence similarly to trypsinized cells.

In addition, one BM-MSC line was selected for the analysis of ΔΨm.

Trypsin- and pronase- detached cells exhibited identical 5,5’,6,6’-tetrachloro-

1,1’,3,3’ tetraethylbenzimidazolcarbocyanine iodide (JC-1) staining patterns, and

most of the cells showed energized or partly depolarized mitochondria compared

with control (protonophore carbonyl cyanide m-chlorophenylhydrazone-treated

cells exhibited fully depolarized mitochondria as expected). Accordingly, this data

indicates that the use of pronase as a substitute for trypsin detachment does not

compromise the viability of MSCs.

5.3.1 The effect of pronase detachment on cell-surface protein epitopes

For 19 cell-surface proteins, pronase treatment decreased the abundance of

corresponding peptide features compared with trypsin treatment (e.g., CD166,

CD49e, CD44, CSPG4, CD29, CD105, CD49c, CD147, CD146, CD99, and

CD90). Also, secreted galectin-1 was digested by pronase. CD29, CD49c, and

CD59 had many novel, pronase-generated peptides, thus revealing a characteristic

cleavage pattern of pronase. Unfortunately, it was not possible to determine in

detail how the cleavage of these peptides affected the actual structure of the

proteins. Also, some peptides from cytosolic proteins had diminished peptide

feature abundance after the pronase detachment, which is probably due to the

cleavage of their interaction partner on the cell surface.

The flow cytometric analysis supported the MS analysis, as there was a clear

decrease in the binding of CD166, CD49e, CD44, CSPG4, and CD146 antibodies

in the pronase-detached cells compared with trypsin-detached cells. The intensity

of anti-CD105 binding was also slightly decreased. Instead, the cell-surface

Page 97: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

95

expressions of galectin-1, CD29, CD49c, D147, CD59, and CD90 were similar

between trypsin- and pronase- detached cells, although the MS analysis had

indicated cleavage of those proteins by pronase. This discrepancy indicates that

the antibodies could recognize the protein epitopes that were still left on the cell

surface after the pronase detachment. Furthermore, the peptides with

glycosylation or other surface modifications could not be detected with MS.

The pronase cleavage of additional adhesion or migration-related proteins

was studied. Significant FN expression on MSCs has been previously described

(Nystedt et al. 2013), and this was further verified in immunofluorescence

staining of BM-MSCs. Pronase always consistently cleaved FN from the cell

surface. Also, CD49d and CD54 expression was lower on the cell surface after

pronase detachment compared with control cells. Their expression levels varied

slightly, even within the same cell line, most likely reflecting the effect of the

culture conditions on expression. The expression of CD106 and CD184 was low

on MSCs. The expression of CD73 was similar in both samples.

The recovery of cleaved cell-surface proteins was studied by flow cytometry.

CD44, CD49e, CSPG4, and CD166 showed almost full recovery after a 7-hour

recovery period. Recovery of proteins was also seen in the MS peptide data for

both intracellular and membrane-bound proteins.

5.3.2 MSC Functionality

To study the effect of pronase on the multipotency or functional properties of

MSCs, the differentiation capacity, immunosuppressive properties, and

angiogenic potential of pronase-detached cells were compared with those of

control cells.

Differentiation

Pronase detachment did not affect the multipotency of MSCs, since both

adipogenic and osteogenic differentiation was successful and comparable to

results after trypsin detachment.

Immunosuppression

Based on the T-cell proliferation assay, the pronase-detached cells inhibited the

proliferation of activated T cells in a dose-dependent manner and just as

Page 98: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

96

effectively as the trypsin-detached cells. This suggests that pronase detachment

does not weaken the immunosuppressive properties of MSCs. T-cell proliferation

was tested with two UCB-MSC lines against allogeneic PBMNCs from two

donors.

Angiogenesis

Both trypsin- and pronase-detached UCB-MSCs were able to strongly stimulate

angiogenesis in the in vitro assay, both directly after detachment and after a 5-

hour recovery. UCB-MSCs required their own culture medium to stimulate tubule

formation. The medium alone was also favourable for angiogenesis, as it

contained growth factors (e.g., platelet-derived growth factor-BB, which is a mild

inducer in this assay). In the test medium, the angiogenic effect of MSCs

remained rather weak. Furthermore, the angiogenic effect was clearly increased

when MSCs were grown in contact with the angiogenesis assay cells compared

with MSCs cultured in inserts.

In Vitro Rolling Assay

Contrary to other functional assays, the pronase detachment had a substantial

impact on BM-MSC adherence and rolling speed in an in vitro flow perfusion

assay. The number of cells having a rolling speed of greater than 220 μm/second

(fast rollers) was clearly higher (p = .056 with t test) in the pronase group (n =35;

mean rolling speed, 261 μm/second) compared with the trypsin group (n = 19;

mean, 221 μm/second). No difference was observed between the groups for the

slowly moving cells (slow rollers).

5.3.3 MSCs and lung clearance

The biodistribution of radioactively labelled BM-MSCs and UCB-MSCs was

studied in mice. More than 75% of the total radioactivity was detected in the

lungs in both cell detachment groups and for both cell types 1 hour after injection.

The rest of the activity was detected in the liver, kidneys, and GI tract, followed

by low radioactivity in spleen, pancreas, heart, and femur. At 15 hours post-

injection, most of the radioactivity was detected in the liver in both cell

detachment groups and for both cell types (49% in the BM-MSC control group

vs. 70% in the BM-MSC pronase group, p = 0.001). There was, however, a

Page 99: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

97

significant difference in total radioactivity measured in the lungs at this time

point, with 32% for control BM-MSCs versus 7% for pronase BM-MSCs (p =

0.001). The amount of radioactivity in the lungs was per se lower for control

UCB-MSCs (3.6%), but in line with the BM-MSCs, as there was clearly less

radioactivity in the lungs for pronase UCB-MSCs (0.6%). Radioactivity was low

in all other organs studied, but generally slightly more radioactivity was observed

when using pronase-detached cells.

The reduced lung retention for pronase-detached cells was confirmed with

fluorescently labeled UCB-MSCs. Twenty hours post-injection, there were one-

fifth as many cells in the lungs in the pronase group than in the control group (p =

0.01), consistent with the results observed using the radioactively labelled cells.

Interestingly, the number of pronase-detached cells was higher in the spleen and

peripheral blood compared with control cells 1 hour post-injection (p = 0.001 and

p = 0.05, respectively). Pronase detachment had no visible effect on cell numbers

in spleen, BM, or blood 20 hours post-injection. Unfortunately, it was not possible

to analyze the liver with this method because of its high enzymatic activity.

Finally, the effect of pronase detachment on lung clearance was verified in a

large-animal model. Consistent with the mouse data, a significant reduction of

lung retention was observed for pronase- detached BM-MSCs in the homologous

porcine model (p= 0.05). In line with the rodent data, prominent radioactivity was

detected in the lungs, kidneys, liver, and spleen in both cell detachment groups

after the 8-hour follow-up. There was, however, more variance in the

biodistribution of the cells in the large animals compared with the mouse model.

No adverse effects due to cell injections were observed, and the porcine groups

had comparable hemodynamic and biochemical data. The lung CT scans revealed

perioperative atelectasis in some individuals in both groups. The effect of the

potentially damaged areas was evaluated, and it was concluded that the atelectasis

did not significantly impact the results.

5.3.4 MSC targeting to an area of injury

The therapeutic advantage of pronase detachment was further studied in a rat

model of carrageenan-induced inflammation. Remarkably, pronase-detached rat

MSCs migrated significantly more effectively to the inflamed area compared with

control cells (p = 0.001). Twice as much radioactivity was detected in the

inflamed paw in the pronase-group compared with the trypsin-group both 50

minutes (0.25 vs. 0.13%, respectively) and 3 hours (0.22 vs. 0.11%) post-

Page 100: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

98

injection. In line with the previous observations made in mice, significantly (p =

0.01) less radioactivity was detected in the lungs of the rats receiving pronase-

detached MSCs at all time-points studied.

Page 101: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

99

6 Discussion

6.1 Biodistribution after intravascular transplantation

The biodistribution pattern seen in Studies I, II and III is consistent with earlier

studies. A common pattern consists of strong lung deposition followed by lower,

but detectable, liver and spleen accumulation (Allers et al. 2004, Barbash et al. 2003, Barbosa da Fonseca et al. 2010, Chin et al. 2003, Detante et al. 2009,

Kraitchman et al. 2005, Makela et al. 2013, Vasconcelos-dos-Santos et al. 2012).

Commonly, the renal accumulation varies across studies. So far, all previous

studies have mainly focused on a specific organ as a therapeutic target and

investigators, including ourselves, have additionally reported overall

biodistribution findings. This lack of unbiased biodistribution studies without any

major ischemic event or other injury led us to conduct our current study with a

porcine model.

Strong BM-MNC homing to bone was observed in Study I. The BM-MNCs

are a mixture of different bone marrow lineage cells and it is possible that

different cell types are responsible for active accumulation within various organs.

BM-MNCs contain haematopoietic cells (HSCs) that are known to home to bone

marrow stem cell niches after administration (Nilsson et al. 1997). Interactions

between HSCs and BM-MSCs have been shown to occur in stem cell niches

(Mendez-Ferrer et al. 2010) and this phenomenon could affect each cell type’s

biological potency in different forms of restorative therapies in as yet unknown

ways. Other major differences in cell biodistribution, when comparing cell

material, were seen in the pattern of migration to the liver, the spleen and the

kidneys. The liver and spleen act as a blood cell reservoirs and so it could be

hypothesized that labelled white blood cells that form a major portion of BM-

MNC population, naturally migrate to these organs. It must also be considered

that the increased liver and spleen accumulation observed in our study after intra-

arterial administration could decrease cellular therapy effectiveness similarly as

lung accumulation – especially so, because we don’t know the exact cell

population that is responsible for the therapeutic effect. BM-MSC administration

resulted in higher activity in the kidneys; it can hypothesised that activity

measured from kidneys can either be a result of real cell migration or homing or

alternatively, Tc99m-HMPAO metabolism. This radiolabel is known to be secreted

via the kidneys, which is confirmed by visualisation of the urinary bladder.

Page 102: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

100

6.2 Lung entrapment

Prominent lung entrapment was present in our porcine specimens in Study I, II

and III, as well as in murine and rat models used in Study III. Study II did not

reveal any obvious differences in lung deposition as a function of follow-up time.

This observation was in contrast to other works reporting diminishing lung

deposition (Chin et al. 2003, Kraitchman et al. 2005, Nystedt et al. 2013).

MSC lung retention may arise from passive entrapment of cells in small-

diameter lung capillaries (Schrepfer et al. 2007, Vilalta et al. 2008) or may be

related to an adhesion molecule-mediated phenomenon (Fischer et al. 2009, Karp

& Leng Teo 2009), as the cell-surface structures appear to at least partially

explain MSC lung retention and clearance (Nystedt et al. 2013).

Whatever the cause for lung adherence, it is obvious that this leads to

diminished biodistribution to other target organs (Fischer et al. 2009, Schrepfer et al. 2007). Intravenously injected cells always encounter the lung capillaries

before reaching the systemic arterial circulation, which leads to the natural

assumption that more cells are found in the lungs after IV administration. An

earlier study has shown that this lung entrapment might even lead to extravasation

and engraftment (Nystedt et al. 2013). We find it interesting that when injecting

the cells intra-arterially, the cells reach the lung after just a single pass through the

circulatory system and still major differences in their organ distribution can be

observed. It could be hypothesised that the cells are sequestered effectively and

specifically by the organ they primarily reach. If this uptake is specific, it might

lead to a change in the freely circulating cell population, especially in the case of

BM-MNCs. Direct cell migration to any target organ can be assumed to be

required for the stem cell paracrine effects (Ranganath et al. 2012). Thus, it could

be crucial that the cells are targeted as accurately as possible to the organ’s

vasculature.

A study by Lee et al. with TSG-6-mediated cardioprotection by hMSCs

entrapped in the lungs is a particularly interesting observation (Lee et al. 2009c).

These results imply that the therapeutic effects of the transplanted MSCs are at

least partially mediated by the portion of the cells trapped in the lungs. Studies

also imply that triggering of IBMIR may be needed for MSC paracrine effects

(Moll et al. 2012). It is still unclear whether decreasing the portion of the cells

deposited in the lungs could have some adverse effects on the outcome of the

cellular therapy. Future studies must compare the actual therapeutic effects with

natural or enhanced lung clearance-pronase detachment methods. Such methods,

Page 103: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

101

which were used in Study III, would offer the opportunity for this comparison, as

we showed increased targeting of the cells to the damaged area after increased

lung clearance.

6.3 The effect of transplantation route on biodistribution

Study I demonstrates that the administration route and cell lineage have major

effects on the acute biodistribution of the transplanted bone marrow-derived cells.

Intra-arterial administration effectively decreases lung accumulation of

transplanted BM-MSCs and appeared to increase their proportion in the liver, the

spleen and the kidneys. When using BM-MNCs, similar variations of

biodistribution could also be observed, but the effect was not as distinct as seen

with the BM-MSCs. The BM-MSCs had a tendency to deposit in the lungs in

larger numbers compared to the BM-MNCs.

Vasconcelos-dos-Santos et al. and also Detante et al. compared BM-MSC

biodistribution differences between IV and IA routes in a rat model of cerebral

ischemia and both reported early lung accumulation and subsequent clearance

with very similar biodistribution to other organs (Detante et al. 2009,

Vasconcelos-dos-Santos et al. 2012). MSC biodistribution was also investigated

in humans. Rosado-de-Castro et al. conducted a biodistribution study on stroke

patients using delayed administration of BM-MNCs through IV or IA routes

(Rosado-de-Castro et al. 2013). They reported lower lung uptake and higher liver

and spleen uptake at the 2 hour time point when using the IA route. Similarly to

animal studies, the high lung uptake of the IV group diminished during the 24

hour follow-up and liver and spleen uptake increased. All these results are

confirmed by our data, which also shows that intra-arterial administration results

in lesser cell accumulation to the lung. Intra-arterial transplantation has also been

suggested as having more therapeutic potency than an intravenous transplantation

(Kamiya et al. 2008). Our observed BM-MNC distribution after the injection in

Studies I and II was comparable with other studies using an intra-arterial

transplantation setup (Correa et al. 2007, Makela et al. 2009).

6.4 Safety assessment

The lung manifestations seen in Study I were most likely due to atelectasis caused

by prolonged mechanical ventilation and the use of muscle relaxants, as both are

rather common phenomenon in clinical practice. Increase of labelled cells in the

Page 104: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

102

lung tissue consolidations could easily explain the activity peaks observed in this

study setup. Both cell lineages have been proven to migrate to injured lung tissue

(Araujo et al. 2010, Rojas et al. 2005). Study I did not reveal any evidence of

pulmonary embolism, as assessed by the contrast enhanced CT. Radiological

diagnostics has its intrinsic limitations and after observance of pulmonary

consolidations, natural concerns of safety arose, as microembolus formation has

also been reported (Furlani et al. 2009, Toma et al. 2009). Possible microemboli

at the subsegmental level cannot be totally excluded with CT-angiography, but the

method’s accuracy should reveal any life threatening emboli.

Recently, it has been suggested that tissue factor could activate thrombotic

events, which has been reported in one case to have led to the death of an

individual who received adipose tissue MSCs (Tatsumi et al. 2013). Pulmonary

hypertension, a classic haemodynamic manifestation of pulmonary embolism,

when physiologic abnormalities of pulmonary embolism patients are assessed

(McIntyre & Sasahara 1971), could not be observed in any animal in Study I. The

porcine, murine and rat models of Studies II and III also did not reveal any

obvious safety issues associated with transplantation of UCBs or bone marrow

MSCs. In addition, other clinical symptoms of pulmonary embolism, such as

decreased arterial oxygen tension, decreased cardiac output or systemic

hypotension, were absent. CT-angiography did not show embolus formation in

the safety controls as well. It remains to be elucidated whether the embolus issue

is only limited to adipose tissue-derived cells, as has already been demonstrated

in mice (Furlani et al. 2009). To the best of our knowledge, there are no reported

adverse effects from the use of BM-MSCs or MNCs. This issue should be

investigated further, but theoretically adipose tissue might be more prone to have

active tissue factor, as it represents cells from the peripheral tissue and bone

marrow cells that also naturally co-exist with blood in the bone marrow.

In Study I, we found notably more lung accumulation of MSCs in the

animals with IV administration. It is likely that the administered cells scatter

throughout the lungs, thereby occluding the microvasculature, as described in

previous studies (Freyman et al. 2006, Furlani et al. 2009, Toma et al. 2009,

Vulliet et al. 2004). Instead of evaluating whether this microembolization occurs,

it is even more important to evaluate the impact on haemodynamics by measuring

increased pulmonary vascular resistance and right ventricular overload, which

could lead to circulatory collapse. Such an effect was not present in our study, and

a similar central venous route for BM-MNC administration was used in a human

trial with pediatric patients suffering from traumatic brain injury (Cox et al.

Page 105: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

103

2011), with no adverse effects being reported. Intravenous transplantation of

autologous MSCs for patients after myocardial infarction also did not reveal any

safety threats (Hare et al. 2009) and similar observations were seen in a study

with ischemic stroke (Lee et al. 2010b).

Rigorous attempts to reveal possible safety threats concerning the

intravascular MSC and BM-MNC transplantation are required. A realistic

approach is essential and possible safety factors should be assessed without

deprecating the possible hazards to ensure patient safety, but exaggerating the

possible adverse effects should also be avoided, as the therapeutic benefit of

cellular therapy could exceed them. When translating the data gained from

preclinical settings to clinical practice, the clinician should be aware of the

rationale for intravascular MSC transplantation to fully capitalise on the

therapeutic potential of these cells in a controlled way.

6.5 IA-transplanted BM-MNCs’ lack of homing to ischemic brain

In Study II, we were unable to detect any significant cerebral migration, which is

in conflict to earlier rodent studies (Li et al. 2001, Correa et al. 2007, Barbosa da

Fonseca et al. 2009, Barbosa da Fonseca et al. 2010). The number of other studies

reporting a negative result is small. No selective administration or focal ischemia

was used, which could explain the differences in the results. In a porcine model

this, however, would require overcoming problems related to selective

intravascular embolization, as the rete mirabilis arterial maze practically prevents

efforts of cerebrovascular cathetisation (Burbridge et al. 2004).

Before conducting Study II, we experimented on a porcine bilateral common

carotid artery occlusion model, which appeared to cause global ischemia

unreliably, and resulted in no alteration in EEG activity. This finding was

hypothesised to originate from circulatory anastomoses and led us to clamp both

the left subclavian artery and the brachiocephalic trunk. The chosen method

resulted in rapid BSR silencing followed by continuous attenuation and slow

recovery. Decreased BSR was associated with ischemic injury in previous studies

(Pokela et al. 2003). We attempted to produce a simplified and reproducible

model of global ischemia with limited biasing factors for the purpose of primarily

observing the behaviour of the transplanted BM-MNCs.

Systematically low results compared with expected migration levels resulted

in discontinuing Study II, as the lack of migration was evident. Our hypothesis

that no higher migration tendency, or statistically significant differences, would

Page 106: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

104

have been detected in this experimental setup if the study was extended to further

time points or conducted with more animals. Lack of migration has also been

reported in a rodent study with a comparable experimental setup (Park et al. 2011). Our study was not conducted further with healthy control animals due to

ethical reasons, as no significant result could be observed in any of the

experimental animals.

An alternative explanation for the lack of homing in this model is the damage

threshold. In global ischemia, the brain tissue oxygen deprivation is life-

threatening after only a short exposure time. This might be insufficient in causing

the required change in the brain vascular endothelium, which ultimately controls

the final homing. With smaller ischemic exposure, the effect is more chronic and

hence more readily sufficient for endothelial activation without a fatal loss of the

brain function.

6.6 The effect of transplantation timing on homing

Intra-arterial transplantation after a stroke has shown to result in BM-MNC

migration to brain tissue at the 2h time point, but not at 24h in a rodent model

(Barbosa da Fonseca et al. 2010). Other studies investigating the impact of timing

on the transplantation’s therapeutic capacity have also generally suggested the

superiority of early transplantation (Barbash et al. 2003, Chen et al. 2001b, de

Vasconcelos Dos Santos et al. 2010, Iihoshi et al. 2004, Yang et al. 2011). This

led us to focus on observing potential cerebral migration during the first 24 hours

of the ischemic insult. Our study did not reveal any obvious culmination point of

migration. The twelve hour time point has been suggested as the peak of

migration in a rodent study with comparable setup (Keimpema E. et al. 2009

Feb).

An optimal window of transplantation is yet to be determined, but we can

postulate that the migration should have occurred during the follow-up in Study

II. The lack of cerebral migration does not rule out the possible therapeutic

benefits of the transplanted cells, as the involvement of indirect paracrine effects

is documented in various clinical settings (Timmers et al. 2007, Keimpema E. et al. 2009, Lee et al. 2009c,).

Page 107: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

105

6.7 The effect of pronase detachment on MSC lung clearance

Study III results show that MSCs can be detached with pronase without altering

the MSC therapeutic capacity. It appears that pronase detachment transiently

modifies MSC surface adhesion proteins, which significantly accelerates the

clearance of these therapeutic cells from the lungs, as shown with both hMSCs in

a xenogeneic mouse model and also in homologous rat and porcine models. In

addition, increased number of pronase-detached cells could be observed in the

inflamed tissue compared with trypsin- detached cells, which suggests potential

beneficial effects of pronase detachment on targeting of MSCs to areas of injury.

As seen in earlier studies (Allers et al. 2004, Barbash et al. 2003, Fischer et al. 2009, Gao et al. 2001, Meyerrose et al. 2007, Nystedt et al. 2013, Schrepfer et al. 2007, Tolar et al. 2006, Vilalta et al. 2008), a majority of the MSCs accumulated

in lungs in Studies I, II, and III, which makes it even more important to optimise

the IV delivery of the MSCs. Despite these obvious issues, the IV route can be

considered as an optimal method of cell transplantation due to its rapid and non-

invasive nature. IV delivery would also be the most suitable for off-the-shelf cell

therapy products.

Existing evidence supports the role of adhesion molecule-mediated lung

deposition of MSCs (Fischer et al. 2009, Nystedt et al. 2013, Ruster et al. 2006)

and, as observed in Study III, pronase accelerated the lung clearance of MSCs.

This finding suggests a specific mechanism linked to cell-surface protein

modification. Pronase also had an impact on the MSC rolling behaviour in a flow

perfusion assay, which further supports the important role of MSC adhesion

characteristics. The functional effect of pronase detachment on lung clearance

was evident in three different animal models in Study III. Although reduced lung

retention was observed in all models, the initial cell retention was more prominent

in a xenogeneic experimental setting used in Study III.

FN is shown to have a strong impact on the adhesion and migration

characteristics of MSCs (Nystedt et al. 2013). Several FN binding receptors are

expressed on lung endothelium, including integrins VLA-4 (CD49d/CD29) and

VLA-5 (CD49e/CD29) and as the MSCs also express these integrins, they

therefore could be able to bind to FN present in the lung endothelium. VLA-4/FN

interactions have even been observed to be involved in migration of MSCs (Yagi et al. 2010b). The blocking of CD49d on MSCs is shown to increase pulmonary

clearance (Fischer et al. 2009). A similar effect has been observed with MSCs

dissolved in heparin, which is known to bind FN (Deak et al. 2010). Study III

Page 108: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

106

showed that FN and CD49d were cleaved by pronase from the cell surface, and

hence this effect could be responsible for the increased pulmonary clearance of

pronase-detached MSCs.

Additionally, pronase cleaved CD44, as indicated by MS and flow cytometry

data. CD44 is observed to affect leukocyte rolling and adhesion to endothelial

cells (Murai et al. 2004) and to also have an impact on MSC migration and

adhesion to endothelium (Brooke et al. 2008, Zhu et al. 2006). CD44 cleavage

could provide a mechanism of mediating pronase-induced pulmonary clearance.

CD54 and CD166 are also involved in endothelial adhesion of MSCs (Brooke et al. 2008); both proteins are also shown to be cleaved by pronase in Study III. It

can be hypothesized that MSC pulmonary adhesion and increased pulmonary

clearance after pronase detachment is a result of manipulation of a range of

different adhesion molecules.

The number of cells did not continuously increase in the injured area after the

earliest time point post-injection. In fact, there was a decrease in the number of

cells in the paws between 50 minutes and 5 hours post-injection. This was also

evident with trypsinized cells and in the control paws, but it was most notable

with pronase-detached cells in the inflamed paw. Cleavage of adhesion molecules

after enzymatic detachment may play a role in the adherence of MSCs to the

injured area. Nevertheless, the amount of pronase-detached cells in the inflamed

paw was always the highest at all time points studied as compared with the

number of cells detected in the other study groups. The amount of pronase-

detached cells remained stabilized from 5 hours onward, suggesting possible

engraftment to the tissue, which coincided with the recovery of the cell-surface

proteins seen in vitro. The acute nature of carrageenan-induced inflammation can

also affect cell recruitment. There is a strong blood flow to the inflamed paw and

rapid formation of edema in the initiation phase of inflammation with a variety of

cytokines attracting the cells, including MSCs, to the site (Hargreaves et al. 1988,

Morris 2003). It may in fact be advantageous that therapeutic cells are recruited to

the inflammation rapidly along with infiltrating neutrophils. After 5 hours, the

edema started to decrease, and it is possible that the paracrine nature of MSCs, via

secretion of beneficial factors that reduce the inflammation, is partly contributing

to diminishing the edema.

Although we have preliminary data to support a beneficial therapeutic effect

of pronase-detached cells in an inflammatory condition, further studies should

explore this in more detail. The results of this study show that pronase detachment

of MSCs resulted in significantly faster clearance of the cells from the lungs due

Page 109: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

107

to a transiently altered cell surface. This was detected with both BM-MSCs and

UCB-MSCs using both small and large animal models in xenogeneic and

homologous experimental setups. In addition, targeting of inflammation was

significantly improved with pronase detachment in a rat model of peripheral

inflammation, most likely because of increased bioavailability. Beneficial

functional consequences of a transiently modified cell surface by pronase were

also supported by the in vitro rolling assay data. Pronase-detached cells retained

therapeutically relevant functional properties, since both the immunosuppressive

and angiogenic potential was equal to that of control cells in vitro. Furthermore,

pronase detachment did not affect the multipotentiality of MSCs. No adverse

effects due to cell injections were observed in the large-animal model, and the

porcine groups had comparable haemodynamic and biochemical data during the

follow-up. Combining pronase detachment with other methods, such as optimal

culture conditions for migratory properties or adding a tag to induce targeting,

could be one way to further develop MSC preparations.

6.8 Clinical applications

Despite the possible systemic effects that the therapeutic cells possess, the

targeting of MSCs to the site of injury is currently considered relevant for some of

their paracrine and restorative functional effects. Commonly, transplanted MSCs

are deposited in the lungs, liver and spleen, as seen in Studies I, II and III; MSCs

targeting to the site of injury generally appears inaccurate (Barbash et al. 2003,

Chapel et al. 2003, Devine et al. 2003, Sackstein et al. 2008, Karp & Leng Teo

2009). We observed in Study I that MSC administration through IA decreased the

pulmonary entrapment of these cells. IA transplantation of MSCs is more invasive

than the IV route, but technical establishment of an IA transplantation route could

be easily performed using pre-existing facilities, such as angiography equipment

or perioperative administration in surgical settings.

Cardiac surgery-related ischemic brain injury would be a promising candidate

for bone marrow-derived stem cell therapy for a number of reasons. The surgical

operations are commonly elective; the occurrence of brain injury can be predicted

in advance. This allows collection and separation of autologous bone marrow

stem cells and the transplantation can be timed in the very early phase, as it is

precisely known when the brain is exposed to ischemia. This also would allow the

use of cultivated autologous MSCs, which have been suggested to have better

clinical effect than BM-MNCs. Cardiac surgery would also allow intra-arterial

Page 110: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

108

transplantation during the operation with minimal additional risks; higher cell

concentrations to the brain and less lung entrapment could be achieved. No

cerebral migration of the BM-MNCs was present in Study II, but this does not

rule out the possible systemic benefits of the therapeutic cells. Future studies

should focus on investigating the occurrence of functional recovery after cell

administration.

In Study III, we showed that pronase detachment increased pulmonary

clearance of the IV-transplanted MSCs and even further increased their

recruitment at a peripheral site of injury. Although certain systemic effects of

MSCs are recognized, the natural assumption is that the cells should target the site

of injury in order to fulfil their paracrine regenerative function.

Immunomodulatory functions of MSCs are well recognized and could have a

major effect in prevention of ischemia-reperfusion injury. In addition, the pronase

detachment procedure is performed without any additional effort compared to

traditional MSC cultivation with trypsin detachment. Futhermore, we observed

that the mechanism behind the enhanced pulmonary passage is based on transient

cell surface modification and the cell viability and differentiation potential is not

compromised, suggesting that their therapeutic function is preserved in this

process.

IV-transplanted MSCs travel through the lung vasculature which could

function as a filtering mechanism against possible thromboemboli caused by

blood contact of the MSCs. Although IA transplantation is another potential

method of increasing tissue targeting of MSCs, pronase detachment could have a

superior safety profile, as injecting possible emboligenic material into the arterial

vasculature could be avoided.

Pronase detachment could provide a viable method of transplantation

optimisation in such clinical conditions, where tissue targeting of MSCs would be

beneficial; classically, this would mean ischemia-related tissue injury at a specific

focus. Overall, the optimal therapeutic indications for pronase-detached MSCs

would be those that lack effective forms of treatment by other means and which

might otherwise be severe for the patient. The possible applications related to

natural vascular diseases could include stroke therapy, MI and embolization of

visceral or peripheral arteries. Another major application could be traumatic

injuries, including traumatic brain injury. Ischemic brain injury caused by cardiac

surgery could also be an indication of IV-transplanted pronase-detached cells.

Page 111: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

109

6.9 Limitations of the study

Correlations between results gained from animal studies and clinical practice is

always a question of great importance. Generally, it is considered that a large

animal model, such as the pigs used in this study, is well comparable to a human

patient. Pigs are considered especially valid as cardiovascular models. Major

organs and their relationships to each other and body size closely resemble the

human body. However, one particular difference is brain size compared to the

whole body, as the porcine brain forms only 0,5% of its body weight. This can be

considered a major limitation of study II, when assessing cell migration to the

damaged brain.

It has been observed that mechanisms of MSC immunomodulation vary

between different species (Ren et al. 2009) and it is possible that preclinical

studies conducted with animals hold potential for bias when translating their

results to human clinical trials. It is commonly recognized that animal studies

employing more highly developed species, such as primates, dogs or pigs, have

closer relevance to human subjects. Comparability between human and porcine

MSCs and their biological properties has been studied by Noort et al. and the

authors have concluded that studies with porcine MSCs can be reliably

extrapolated to correspond with results that would be seen when using human

MSCs (Noort et al. 2012). Our results with a porcine model cannot be considered

as directly translatable to human trials, but general information provided by

Studies II and III can be considered accurate enough to act as guidance for future

trials.

The Tc-HMPAO labelling studies offer the potential to globally track the

biodistribution of the transplanted cells in a porcine model. This method,

however, has its limitations due to the limited half-life of the radionuclide isotope.

Obvious limitations of Studies I, II and III is the small sample size. The variation

seen in these studies is indicative of the need for power calculations for further

studies.

Another limitation of Study I is related to the difference in the cell types used.

Cell doses were not directly comparable, as a tenfold greater quantity of BM-

MNCs was used. This was required to reach similar input radioactivity doses, as

the labelling efficiency of BM-MSCs was also tenfold higher. The applied BM-

MSC quantity was comparable to a porcine study conducted by Wolf et al. in

which the dose effectiveness of BM-MSCs was examined in a myocardial

infarction model (Wolf et al. 2009). Although organ accumulation trends reported

Page 112: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

110

in Study I can be compared between cell lineages after normalization, results

should be interpreted with caution, as a tenfold higher dose presumably affects

administration dynamics; larger cell doses are reported to cause more capillary

occlusion (Furlani et al. 2009). Furthermore, BM-MNC groups underwent bone

marrow harvesting, while the BM-MSC groups did not, which could bias the

presumed native biodistribution due to local mechanical disruption of the bone

marrow.

6.10 Summary

In Study I, our data indicates that intra-arterial administration appears to possess

the potential to allow more cells to reach the systemic vasculature, thereby

targeting organs of cellular therapy, such as the brain, heart and kidneys, whereas

intra-venous administration allows targeting of the lung, which can also be

clinically relevant. None of the administration combinations showed any evidence

of safety threats, but an awareness of the possibility of embolization should result

in a cautious approach to utilising intravascular cell administration. Although this

experimental model is limited to offering a descriptive insight on the subject,

future studies could provide basic mapping of BM-MSC and BM-MNC

biodistributions following systemic IA or IV administration.

In contrast to promising earlier studies on acute focal ischemia, the

transplantation of BM-MNCs in Study II did not result in the retention of cells in

the damaged brain in the porcine global ischemia model. BM-MNCs did not

migrate into brain tissue in substantial numbers during first 24 hours, as the cells

accumulated in other major organs and the bone marrow.

Study III showed that pronase detachment could be used to improve the MSC

lung clearance and targeting in vivo without compromising the therapeutic

properties of the cells. Pronase detachment could be easily incorporated into the

clinical production processes of adherent cells, since it could replace trypsin.

However, there are still many questions surrounding the clinical use of pronase,

including the safety of the product and the demonstration of the actual

improvement of functional outcome.

Page 113: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

111

7 Conclusion

I Both the BM-MNCs and MSCs deposit mainly in the lungs, liver and spleen.

IA administration of MSCs diminishes their lung deposition. Intravascular

transplantation of these cells did not cause pulmonary emboli.

II Intra-arterially transplanted BM-MNCs do not reach the brain in significant

numbers in a porcine model of global ischemic-induced brain injury and there

is not a culmination point of migration during the first 24 hours

III Transient proteolytic modification of MSCs with pronase decreased lung

accumulation and improved migration to the site of injury without affecting

the characteristics of the cells.

Page 114: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

112

Page 115: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

113

References

Abbott JD, Huang Y, Liu D, Hickey R, Krause DS & Giordano FJ (2004) Stromal Cell–Derived Factor-1α Plays a Critical Role in Stem Cell Recruitment to the Heart After Myocardial Infarction but Is Not Sufficient to Induce Homing in the Absence of Injury. Circulation 110(21): 3300–3305.

Aggarwal S & Pittenger MF (2005) Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 105(4): 1815–1822.

Aicher A, Brenner W, Zuhayra M, Badorff C, Massoudi S, Assmus B, Eckey T, Henze E, Zeiher AM & Dimmeler S (2003) Assessment of the tissue distribution of transplanted human endothelial progenitor cells by radioactive labeling. Circulation 107(16): 2134–2139.

Allers C, Sierralta WD, Neubauer S, Rivera F, Minguell JJ & Conget PA (2004) Dynamic of distribution of human bone marrow-derived mesenchymal stem cells after transplantation into adult unconditioned mice. Transplantation 78(4): 503-508.

Anjos-Afonso F, Siapati EK & Bonnet D (2004) In vivo contribution of murine mesenchymal stem cells into multiple cell-types under minimal damage conditions. J Cell Sci 117(Pt 23): 5655–5664.

Araujo IM, Abreu SC, Maron-Gutierrez T, Cruz F, Fujisaki L, Carreira H,Jr, Ornellas F, Ornellas D, Vieira-de-Abreu A, Castro-Faria-Neto HC, Muxfeldt Ab'Saber A, Teodoro WR, Diaz BL, Peres Dacosta C, Capelozzi VL, Pelosi P, Morales MM & Rocco PR (2010) Bone marrow-derived mononuclear cell therapy in experimental pulmonary and extrapulmonary acute lung injury. Crit Care Med 38(8): 1733–1741.

Arvidson K, Abdallah BM, Applegate LA, Baldini N, Cenni E, Gomez-Barrena E, Granchi D, Kassem M, Konttinen YT, Mustafa K, Pioletti DP, Sillat T & Finne-Wistrand A (2011) Bone regeneration and stem cells. J Cell Mol Med 15(4): 718–746.

Augello A, Tasso R, Negrini SM, Amateis A, Indiveri F, Cancedda R & Pennesi G (2005) Bone marrow mesenchymal progenitor cells inhibit lymphocyte proliferation by activation of the programmed death 1 pathway. Eur J Immunol 35(5): 1482–1490.

Bantubungi K, Blum D, Cuvelier L, Wislet-Gendebien S, Rogister B, Brouillet E & Schiffmann SN (2008) Stem cell factor and mesenchymal and neural stem cell transplantation in a rat model of Huntington's disease. Mol Cell Neurosci 37(3): 454–470.

Bao X, Wei J, Feng M, Lu S, Li G, Dou W, Ma W, Ma S, An Y, Qin C, Zhao RC & Wang R (2010) Transplantation of human bone marrow-derived mesenchymal stem cells promotes behavioral recovery and endogenous neurogenesis after cerebral ischemia in rats. Brain Res 1367: 103–113.

Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion S, Tessone A, Miller L, Guetta E, Zipori D, Kedes LH, Kloner RA & Leor J (2003) Systemic Delivery of Bone Marrow–Derived Mesenchymal Stem Cells to the Infarcted Myocardium. Circulation 108(7): 863–868.

Page 116: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

114

Barbosa da Fonseca LM, Gutfilen B, Rosado de Castro PH, Battistella V, Goldenberg RC, Kasai-Brunswick T, Chagas CL, Wajnberg E, Maiolino A, Salles Xavier S, Andre C, Mendez-Otero R & de Freitas GR (2010) Migration and homing of bone-marrow mononuclear cells in chronic ischemic stroke after intra-arterial injection. Exp Neurol 221(1): 122–128.

Barbosa da Fonseca LM, Battistella V, de Freitas GR, Gutfilen B, dos Santos Goldenberg RC, Maiolino A, Wajnberg E, Rosado de Castro PH, Mendez-Otero R & Andre C (2009) Early Tissue Distribution of Bone Marrow Mononuclear Cells After Intra-Arterial Delivery in a Patient With Chronic Stroke. Circulation 120(6): 539–541.

Barkholt L, Flory E, Jekerle V, Lucas-Samuel S, Ahnert P, Bisset L, Büscher D, Fibbe W, Foussat A, Kwa M, Lantz O, Mačiulaitis R, Palomäki T, Schneider CK, Sensebé L, Tachdjian G, Tarte K, Tosca L & Salmikangas P (2013). Risk of tumorigenicity in mesenchymal stromal cell-based therapies–bridging scientific observations and regulatory viewpoints. Cytotherapy 15(7):753–9.

Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, Hardy W, Devine S, Ucker D, Deans R, Moseley A & Hoffman R (2002) Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 30(1): 42–48.

Becker AJ, McCulloch EA & Till JE (1963) Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature 197: 452–454.

Beyth S, Borovsky Z, Mevorach D, Liebergall M, Gazit Z, Aslan H, Galun E & Rachmilewitz J (2005) Human mesenchymal stem cells alter antigen-presenting cell maturation and induce T-cell unresponsiveness. Blood 105(5): 2214–2219.

Bhakta S, Hong P & Koc O (2006) The surface adhesion molecule CXCR4 stimulates mesenchymal stem cell migration to stromal cell-derived factor-1 in vitro but does not decrease apoptosis under serum deprivation. Cardiovasc Revasc Med 7(1): 19–24.

Borlongan CV, Lind JG, Dillon-Carter O, Yu G, Hadman M, Cheng C, Carroll J & Hess DC (2004) Bone marrow grafts restore cerebral blood flow and blood brain barrier in stroke rats. Brain Res 1010(1-2): 108–116.

Bossolasco P, Cova L, Calzarossa C, Rimoldi SG, Borsotti C, Deliliers GL, Silani V, Soligo D & Polli E (2005) Neuro-glial differentiation of human bone marrow stem cells in vitro. Exp Neurol 193(2): 312–325.

Breitbach M, Bostani T, Roell W, Xia Y, Dewald O, Nygren JM, Fries JW, Tiemann K, Bohlen H, Hescheler J, Welz A, Bloch W, Jacobsen SE & Fleischmann BK (2007) Potential risks of bone marrow cell transplantation into infarcted hearts. Blood 110(4): 1362–1369.

Brooke G, Tong H, Levesque JP & Atkinson K (2008) Molecular trafficking mechanisms of multipotent mesenchymal stem cells derived from human bone marrow and placenta. Stem Cells Dev 17(5): 929–940.

Burbridge B, Matte G & Remedios A (2004) Complex intracranial arterial anatomy in swine is unsuitable for cerebral infarction projects. Can Assoc Radiol J 55(5): 326-329.

Caplan AI (1991) Mesenchymal stem cells. J Orthop Res 9(5): 641–650.

Page 117: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

115

Chapel A, Bertho JM, Bensidhoum M, Fouillard L, Young RG, Frick J, Demarquay C, Cuvelier F, Mathieu E, Trompier F, Dudoignon N, Germain C, Mazurier C, Aigueperse J, Borneman J, Gorin NC, Gourmelon P & Thierry D (2003) Mesenchymal stem cells home to injured tissues when co-infused with hematopoietic cells to treat a radiation-induced multi-organ failure syndrome. J Gene Med 5(12): 1028–1038.

Chen J, Li Y & Chopp M (2000) Intracerebral transplantation of bone marrow with BDNF after MCAo in rat. Neuropharmacology 39(5): 711–716.

Chen J, Li Y, Wang L, Lu M, Zhang X & Chopp M (2001a) Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats. J Neurol Sci 189(1-2): 49–57.

Chen J, Li Y, Wang L, Zhang Z, Lu D, Lu M & Chopp M (2001b) Therapeutic Benefit of Intravenous Administration of Bone Marrow Stromal Cells After Cerebral Ischemia in Rats. Stroke 32(4): 1005–1011.

Chen J, Zhang ZG, Li Y, Wang L, Xu YX, Gautam SC, Lu M, Zhu Z & Chopp M (2003) Intravenous Administration of Human Bone Marrow Stromal Cells Induces Angiogenesis in the Ischemic Boundary Zone After Stroke in Rats. Circulation Research 92(6): 692–699.

Chen X, Li Y, Wang L, Katakowski M, Zhang L, Chen J, Xu Y, Gautam SC & Chopp M (2002) Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology 22(4): 275–279.

Cheng H, Byrska-Bishop M, Zhang CT, Kastrup CJ, Hwang NS, Tai AK, Lee WW, Xu X, Nahrendorf M, Langer R & Anderson DG (2012) Stem cell membrane engineering for cell rolling using peptide conjugation and tuning of cell-selectin interaction kinetics. Biomaterials 33(20): 5004–5012.

Chin BB, Nakamoto Y, Bulte JW, Pittenger MF, Wahl R & Kraitchman DL (2003) 111In oxine labelled mesenchymal stem cell SPECT after intravenous administration in myocardial infarction. Nucl Med Commun 24(11): 1149–1154.

Chopp M & Li Y (2002) Treatment of neural injury with marrow stromal cells. The Lancet Neurology 1(2): 92–100.

Correa PL, Mesquita CT, Felix RM, Azevedo JC, Barbirato GB, Falcao CH, Gonzalez C, Mendonca ML, Manfrim A, de Freitas G, Oliveira CC, Silva D, Avila D, Borojevic R, Alves S, Oliveira AC,Jr & Dohmann HF (2007) Assessment of intra-arterial injected autologous bone marrow mononuclear cell distribution by radioactive labeling in acute ischemic stroke. Clin Nucl Med 32(11): 839–841.

Cox CS Jr, Baumgartner JE, Harting MT, Worth LL, Walker PA, Shah SK, Ewing-Cobbs L, Hasan KM, Day MC, Lee D, Jimenez F & Gee A (2011) Autologous bone marrow mononuclear cell therapy for severe traumatic brain injury in children. Neurosurgery 68(3): 588–600.

Coyne TM, Marcus AJ, Woodbury D & Black IB (2006) Marrow stromal cells transplanted to the adult brain are rejected by an inflammatory response and transfer donor labels to host neurons and glia. Stem Cells 24(11): 2483–2492.

Page 118: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

116

Crigler L, Robey RC, Asawachaicharn A, Gaupp D & Phinney DG (2006) Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp Neurol 198(1): 54–64.

Cuende N, Rico L & Herrera C (2012). Concise review: bone marrow mononuclear cells for the treatment of ischemic syndromes: medicinal product or cell transplantation? Stem Cells Transl Med. 1(5):403–8

De Bari C, Dell'Accio F, Tylzanowski P & Luyten FP (2001) Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum 44(8): 1928–1942.

de Vasconcelos Dos Santos A, da Costa Reis J, Diaz Paredes B, Moraes L, Jasmin, Giraldi-Guimaraes A & Mendez-Otero R (2010) Therapeutic window for treatment of cortical ischemia with bone marrow-derived cells in rats. Brain Res 1306: 149–158.

Deak E, Ruster B, Keller L, Eckert K, Fichtner I, Seifried E & Henschler R (2010) Suspension medium influences interaction of mesenchymal stromal cells with endothelium and pulmonary toxicity after transplantation in mice. Cytotherapy 12(2): 260–264.

Deng W, Obrocka M, Fischer I & Prockop DJ (2001) In vitro differentiation of human marrow stromal cells into early progenitors of neural cells by conditions that increase intracellular cyclic AMP. Biochem Biophys Res Commun 282(1): 148–152.

Detante O, Moisan A, Dimastromatteo J, Richard MJ, Riou L, Grillon E, Barbier E, Desruet MD, De Fraipont F, Segebarth C, Jaillard A, Hommel M, Ghezzi C & Remy C (2009) Intravenous administration of 99mTc-HMPAO-labeled human mesenchymal stem cells after stroke: in vivo imaging and biodistribution. Cell Transplant 18(12): 1369–1379.

Devine SM, Cobbs C, Jennings M, Bartholomew A & Hoffman R (2003) Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood 101(8): 2999–3001.

Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S & Gianni AM (2002) Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 99(10): 3838–3843.

Djouad F, Plence P, Bony C, Tropel P, Apparailly F, Sany J, Noel D & Jorgensen C (2003) Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood 102(10): 3837–3844.

Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D & Horwitz E (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement Cytotherapy 8(4): 315–317.

Erices A, Conget P & Minguell JJ (2000) Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 109(1): 235–242.

Fischer UM, Harting MT, Jimenez F, Monzon-Posadas WO, Xue H, Savitz SI, Laine GA & Cox CS Jr (2009) Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary first-pass effect. Stem Cells Dev 18(5): 683–692.

Page 119: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

117

Foley JF & Aftonomos B (1970) The use of pronase in tissue culture: a comparison with trypsin. J Cell Physiol 75(2): 159–161.

Freyman T, Polin G, Osman H, Crary J, Lu M, Cheng L, Palasis M & Wilensky RL (2006) A quantitative, randomized study evaluating three methods of mesenchymal stem cell delivery following myocardial infarction. Eur Heart J 27(9): 1114–1122.

Friedenstein AJ, Chailakhjan RK & Lalykina KS (1970) The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet 3(4): 393–403.

Fujita Y, Ihara M, Ushiki T, Hirai H, Kizaka-Kondoh S, Hiraoka M, Ito H & Takahashi R (2010) Early Protective Effect of Bone Marrow Mononuclear Cells Against Ischemic White Matter Damage Through Augmentation of Cerebral Blood Flow. Stroke 41(12): 2938–2943.

Furlani D, Ugurlucan M, Ong L, Bieback K, Pittermann E, Westien I, Wang W, Yerebakan C, Li W, Gaebel R, Li RK, Vollmar B, Steinhoff G & Ma N (2009) Is the intravascular administration of mesenchymal stem cells safe? Mesenchymal stem cells and intravital microscopy. Microvasc Res 77(3): 370–376.

Galindo LT, Filippo TR, Semedo P, Ariza CB, Moreira CM, Camara NO & Porcionatto MA (2011) Mesenchymal stem cell therapy modulates the inflammatory response in experimental traumatic brain injury. Neurol Res Int 2011: 564089.

Gao J, Dennis JE, Muzic RF, Lundberg M & Caplan AI (2001) The dynamic in vivo distribution of bone marrow-derived mesenchymal stem cells after infusion. Cells Tissues Organs 169(1): 12–20.

Glennie S, Soeiro I, Dyson PJ, Lam EW & Dazzi F (2005) Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood 105(7): 2821–2827.

Goren A, Dahan N, Goren E, Baruch L & Machluf M (2010) Encapsulated human mesenchymal stem cells: a unique hypoimmunogenic platform for long-term cellular therapy. FASEB J 24(1): 22–31.

Gronthos S, Mankani M, Brahim J, Robey PG & Shi S (2000) Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci U S A 97(25): 13625–13630.

Gupta PK, Chullikana A, Parakh R, Desai S, Das A, Gottipamula S, Krishnamurthy S, Anthony N, Pherwani A & Majumdar AS (2013) A double blind randomized placebo controlled phase I/II study assessing the safety and efficacy of allogeneic bone marrow derived mesenchymal stem cell in critical limb ischemia. J Transl Med 11: 143–5876-11-143.

Hannoush EJ, Sifri ZC, Elhassan IO, Mohr AM, Alzate WD, Offin M & Livingston DH (2011) Impact of enhanced mobilization of bone marrow derived cells to site of injury. J Trauma 71(2): 283-9; discussion 289–91.

Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, Gerstenblith G, DeMaria AN, Denktas AE, Gammon RS, Hermiller JB Jr, Reisman MA, Schaer GL & Sherman W (2009) A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol 54(24): 2277–2286.

Page 120: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

118

Hargreaves K, Dubner R, Brown F, Flores C & Joris J (1988) A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 32(1): 77–88.

Harting MT, Jimenez F, Xue H, Fischer UM, Baumgartner J, Dash PK & Cox CS (2009) Intravenous mesenchymal stem cell therapy for traumatic brain injury. J Neurosurg 110(6): 1189–1197.

Hemeda H, Jakob M, Ludwig AK, Giebel B, Lang S & Brandau S (2010) Interferon-gamma and tumor necrosis factor-alpha differentially affect cytokine expression and migration properties of mesenchymal stem cells. Stem Cells Dev 19(5): 693–706.

Hoogduijn MJ, Roemeling-van Rhijn M, Engela AU, Korevaar SS, Mensah FK, Franquesa M, de Bruin RW, Betjes MG, Weimar W & Baan CC (2013) Mesenchymal stem cells induce an inflammatory response after intravenous infusion. Stem Cells Dev 22(21): 2825–2835.

Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, McNall RY, Muul L & 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 U S A. 25;99(13):8932–7.

Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M, Sussman M, Orchard P, Marx JC, Pyeritz RE & Brenner MK (1999) Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med 5(3): 309–313.

Hung SC, Pochampally RR, Hsu SC, Sanchez C, Chen SC, Spees J & Prockop DJ (2007) Short-term exposure of multipotent stromal cells to low oxygen increases their expression of CX3CR1 and CXCR4 and their engraftment in vivo. PLoS One 2(5): e416.

Iihoshi S, Honmou O, Houkin K, Hashi K & Kocsis JD (2004) A therapeutic window for intravenous administration of autologous bone marrow after cerebral ischemia in adult rats. Brain Res 1007(1-2): 1–9.

In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GM, Claas FH, Fibbe WE & Kanhai HH (2004) Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 22(7): 1338–1345.

Inoue M, Honmou O, Oka S, Houkin K, Hashi K & Kocsis JD (2003) Comparative analysis of remyelinating potential of focal and intravenous administration of autologous bone marrow cells into the rat demyelinated spinal cord. Glia 44(2): 111–118.

Ip JE, Wu Y, Huang J, Zhang L, Pratt RE & Dzau VJ (2007) Mesenchymal stem cells use integrin beta1 not CXC chemokine receptor 4 for myocardial migration and engraftment. Mol Biol Cell 18(8): 2873–2882.

Isakova IA, Dufour J, Lanclos C, Bruhn J & Phinney DG (2010) Cell-dose-dependent increases in circulating levels of immune effector cells in rhesus macaques following intracranial injection of allogeneic MSCs. Exp Hematol 38(10): 957–967.e1.

Isele NB, Lee HS, Landshamer S, Straube A, Padovan CS, Plesnila N & Culmsee C (2007) Bone marrow stromal cells mediate protection through stimulation of PI3-K/Akt and MAPK signaling in neurons. Neurochem Int 50(1): 243–250.

Page 121: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

119

Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD & Mao N (2005) Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood 105(10): 4120–4126.

Kaivosoja E, Barreto G, Levon K, Virtanen S, Ainola M & Konttinen YT (2012) Chemical and physical properties of regenerative medicine materials controlling stem cell fate. Ann Med 44(7): 635–650.

Kamiya N, Ueda M, Igarashi H, Nishiyama Y, Suda S, Inaba T & Katayama Y (2008) Intra-arterial transplantation of bone marrow mononuclear cells immediately after reperfusion decreases brain injury after focal ischemia in rats. Life Sci 83(11–12): 433–437.

Karantalis V, DiFede DL, Gerstenblith G, Pham S, Symes J, Zambrano JP, Fishman J, Pattany P, McNiece I, Conte J, Schulman S, Wu K, Shah A, Breton E, Davis-Sproul J, Schwarz R, Feigenbaum G, Mushtaq M, Suncion VY, Lardo AC, Borrello I, Mendizabal A, Karas TZ, Byrnes J, Lowery M, Heldman AW & Hare JM (2014) Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The Prospective Randomized Study of Mesenchymal Stem Cell Therapy in Patients Undergoing Cardiac Surgery (PROMETHEUS) trial. Circ Res 11;114(8):1302–10

Karp JM & Leng Teo GS (2009) Mesenchymal stem cell homing: the devil is in the details. Cell Stem Cell 4(3): 206–216.

Kean TJ, Duesler L, Young RG, Dadabayev A, Olenyik A, Penn M, Wagner J, Fink DJ, Caplan AI & Dennis JE (2012) Development of a peptide-targeted, myocardial ischemia-homing, mesenchymal stem cell. J Drug Target 20(1): 23–32.

Keimpema E., Fokkens MR. & Nagy Z, Agoston V, Luiten PG, Nyakas C, Boddeke HW, Copray JC. (2009 Feb) Early transient presence of implanted bone marrow stem cells reduces lesion size after cerebral ischaemia in adult rats. Neuropathol Appl Neurobiol 35(1): 89–102.

Kharaziha P, Hellstrom PM, Noorinayer B, Farzaneh F, Aghajani K, Jafari F, Telkabadi M, Atashi A, Honardoost M, Zali MR & Soleimani M (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(10): 1199–1205.

Koh SH, Kim KS, Choi MR, Jung KH, Park KS, Chai YG, Roh W, Hwang SJ, Ko HJ, Huh YM, Kim HT & Kim SH (2008) Implantation of human umbilical cord-derived mesenchymal stem cells as a neuroprotective therapy for ischemic stroke in rats. Brain Res 1229: 233–248.

Kopen GC, Prockop DJ & Phinney DG (1999) Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proceedings of the National Academy of Sciences 96(19): 10711–10716.

Page 122: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

120

Kraitchman DL, Tatsumi M, Gilson WD, Ishimori T, Kedziorek D, Walczak P, Segars WP, Chen HH, Fritzges D, Izbudak I, Young RG, Marcelino M, Pittenger MF, Solaiyappan M, Boston RC, Tsui BM, Wahl RL & Bulte JW (2005) Dynamic imaging of allogeneic mesenchymal stem cells trafficking to myocardial infarction. Circulation 112(10): 1451–1461.

Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, Andreini A, Santarlasci V, Mazzinghi B, Pizzolo G, Vinante F, Romagnani P, Maggi E, Romagnani S & Annunziato F (2006) Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 24(2): 386–398.

Lager KE, Mistri AK, Khunti K, Haunton VJ, Sett AK & Wilson AD (2014). Interventions for improving modifiable risk factor control in the secondary prevention of stroke. Cochrane Database Syst Rev. May 2;5:CD009103

Laitinen A, Nystedt J & Laitinen S (2011) The isolation and culture of human cord blood-derived mesenchymal stem cells under low oxygen conditions. Methods Mol Biol 698: 63–73.

Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, Lanino E, Sundberg B, Bernardo ME, Remberger M, Dini G, Egeler RM, Bacigalupo A, Fibbe W, Ringden O & Developmental Committee of the European Group for Blood and Marrow Transplantation (2008) Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet 371(9624): 1579–1586.

Le Blanc K, Tammik L, Sundberg B, Haynesworth SE & Ringden O (2003) Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex. Scand J Immunol 57(1): 11–20.

Lee JA, Kim BI, Jo CH, Choi CW, Kim EK, Kim HS, Yoon KS & Choi JH (2010a) Mesenchymal stem-cell transplantation for hypoxic-ischemic brain injury in neonatal rat model. Pediatr Res 67(1): 42–46.

Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY & STARTING collaborators (2010b) A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke Stem Cells 28(6): 1099–1106.

Lee JW, Fang X, Gupta N, Serikov V & Matthay MA (2009a) 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(38): 16357–16362.

Lee JW, Gupta N, Serikov V & Matthay MA (2009b) Potential application of mesenchymal stem cells in acute lung injury. Expert Opin Biol Ther 9(10): 1259–1270.

Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, Semprun-Prieto L, Delafontaine P & Prockop DJ (2009c) Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell 5(1): 54–63.

Page 123: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

121

Lee RH, Seo MJ, Pulin AA, Gregory CA, Ylostalo J & Prockop DJ (2009d) The CD34-like protein PODXL and alpha6-integrin (CD49f) identify early progenitor MSCs with increased clonogenicity and migration to infarcted heart in mice. Blood 113(4): 816–826.

Lehtinen M, Pätilä T, Vento A, Kankuri E, Suojaranta-Ylinen R, Pöyhiä R & Harjula A; for the Helsinki BMMC Collaboration. (2014) Prospective, randomized, double-blinded trial of bone marrow cell transplantation combined with coronary surgery - perioperative safety study. Interact Cardiovasc Thorac Surg. 20

Leskela HV, Risteli J, Niskanen S, Koivunen J, Ivaska KK & Lehenkari P (2003) Osteoblast recruitment from stem cells does not decrease by age at late adulthood. Biochem Biophys Res Commun 311(4): 1008–1013.

Levit RD, Landazuri N, Phelps EA, Brown ME, Garcia AJ, Davis ME, Joseph G, Long R, Safley SA, Suever JD, Lyle AN, Weber CJ & Taylor WR (2013) Cellular encapsulation enhances cardiac repair. J Am Heart Assoc 2(5): e000367.

Li Y, Chen J, Wang L, Lu M & Chopp M (2001) Treatment of stroke in rat with intracarotid administration of marrow stromal cells. Neurology; Neurology 56(12): 1666–1672.

Likosky DS, Leavitt BJ, Marrin CA, Malenka DJ, Reeves AG, Weintraub RM, Caplan LR, Baribeau YR, Charlesworth DC, Ross CS, Braxton JH, Hernandez F Jr, O'Connor GT & Northern New England Cardiovascular Disease Study Group (2003) Intra- and postoperative predictors of stroke after coronary artery bypass grafting. Ann Thorac Surg. 76(2):428–34

Lin CS, Xin ZC, Dai J & Lue TF (2013) Commonly used mesenchymal stem cell markers and tracking labels: Limitations and challenges. Histol Histopathol 28(9): 1109–1116.

Makela J, Anttila V, Ylitalo K, Takalo R, Lehtonen S, Makikallio T, Niemela E, Dahlbacka S, Tikkanen J, Kiviluoma K, Juvonen T & Lehenkari P (2009) Acute homing of bone marrow-derived mononuclear cells in intramyocardial vs. intracoronary transplantation Scand Cardiovasc J 43(6): 366–373.

Makela T, Yannopoulos F, Alestalo K, Makela J, Lepola P, Anttila V, Lehtonen S, Kiviluoma K, Takalo R, Juvonen T & Lehenkari P (2013) Intra-arterial bone marrow mononuclear cell distribution in experimental global brain ischaemia. Scand Cardiovasc J 47(2): 114–120.

Martens TP, See F, Schuster MD, Sondermeijer HP, Hefti MM, Zannettino A, Gronthos S, Seki T & Itescu S (2006) Mesenchymal lineage precursor cells induce vascular network formation in ischemic myocardium. Nat Clin Pract Cardiovasc Med 3 Suppl 1: S18–22.

Mathiasen AB, Jørgensen E, Qayyum AA, Haack-Sørensen M, Ekblond A & Kastrup J (2012) Rationale and design of the first randomized, double-blind, placebo-controlled trial of intramyocardial injection of autologous bone-marrow derived Mesenchymal Stromal Cells in chronic ischemic Heart Failure (MSC-HF Trial). Am Heart J. 164(3):285–91

Page 124: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

122

Mazo M, Gavira JJ, Abizanda G, Moreno C, Ecay M, Soriano M, Aranda P, Collantes M, Alegria E, Merino J, Penuelas I, Garcia Verdugo JM, Pelacho B & Prosper F (2010) Transplantation of mesenchymal stem cells exerts a greater long-term effect than bone marrow mononuclear cells in a chronic myocardial infarction model in rat. Cell Transplant 19(3): 313–328.

McIntyre KM & Sasahara AA (1971) The hemodynamic response to pulmonary embolism in patients without prior cardiopulmonary disease. Am J Cardiol 28(3): 288–294.

Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, Scadden DT, Ma'ayan A, Enikolopov GN & Frenette PS (2010) Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466(7308): 829–834.

Meyerrose TE, De Ugarte DA, Hofling AA, Herrbrich PE, Cordonnier TD, Shultz LD, Eagon JC, Wirthlin L, Sands MS, Hedrick MA & Nolta JA (2007) In vivo distribution of human adipose-derived mesenchymal stem cells in novel xenotransplantation models. Stem Cells 25(1): 220–227.

Mezey E, Chandross KJ, Harta G, Maki RA & McKercher SR (2000) Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow. Science 290(5497): 1779–1782.

Mitkari B, Kerkela E, Nystedt J, Korhonen M, Mikkonen V, Huhtala T & Jolkkonen J (2013) Intra-arterial infusion of human bone marrow-derived mesenchymal stem cells results in transient localization in the brain after cerebral ischemia in rats. Exp Neurol 239: 158–162.

Moll G, Rasmusson-Duprez I, von Bahr L, Connolly-Andersen AM, Elgue G, Funke L, Hamad OA, Lonnies H, Magnusson PU, Sanchez J, Teramura Y, Nilsson-Ekdahl K, Ringden O, Korsgren O, Nilsson B & Le Blanc K (2012) Are therapeutic human mesenchymal stromal cells compatible with human blood? Stem Cells 30(7): 1565–1574.

Moniche F, Gonzalez A, Gonzalez-Marcos J, Carmona M, Piñero P, Espigado I, Garcia-Solis D, Cayuela A, Montaner J, Boada C, Rosell A, Jimenez M, Mayol A & Gil-Peralta A (2012) Intra-Arterial Bone Marrow Mononuclear Cells in Ischemic Stroke. Stroke 43(8): 2242–2244.

Morris CJ (2003) Carrageenan-induced paw edema in the rat and mouse. Methods Mol Biol 225: 115–121.

Morrison SJ & Scadden DT (2014) The bone marrow niche for haematopoietic stem cells. Nature 16;505(7483):327–34

Murai T, Sougawa N, Kawashima H, Yamaguchi K & Miyasaka M (2004) CD44-chondroitin sulfate interactions mediate leukocyte rolling under physiological flow conditions. Immunol Lett 93(2-3): 163–170.

Nadri S & Soleimani M (2007) Comparative analysis of mesenchymal stromal cells from murine bone marrow and amniotic fluid. Cytotherapy 9(8): 729–737.

Nilsson SK, Dooner MS, Tiarks CY, Weier HU & Quesenberry PJ (1997) Potential and distribution of transplanted hematopoietic stem cells in a nonablated mouse model. Blood 89(11): 4013–4020.

Page 125: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

123

Noort WA, Oerlemans MI, Rozemuller H, Feyen D, Jaksani S, Stecher D, Naaijkens B, Martens AC, Buhring HJ, Doevendans PA & Sluijter JP (2012) Human versus porcine mesenchymal stromal cells: phenotype, differentiation potential, immunomodulation and cardiac improvement after transplantation. J Cell Mol Med 16(8): 1827–1839.

Nystedt J, Anderson H, Tikkanen J, Pietila M, Hirvonen T, Takalo R, Heiskanen A, Satomaa T, Natunen S, Lehtonen S, Hakkarainen T, Korhonen M, Laitinen S, Valmu L & Lehenkari P (2013) Cell surface structures influence lung clearance rate of systemically infused mesenchymal stromal cells. Stem Cells 31(2): 317–326.

Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM & Leri A, Anversa P (2001) Bone marrow cells regenerate infarcted myocardium. Nature 5;410(6829):701–5

Parekkadan B, Tilles AW & Yarmush ML (2008) Bone marrow-derived mesenchymal stem cells ameliorate autoimmune enteropathy independently of regulatory T cells. Stem Cells 26(7): 1913–1919.

Park B, Shim W, Lee G, Bang OY, An Y, Yoon J & Ahn YH (2011) Early distribution of intravenously injected mesenchymal stem cells in rats with acute brain trauma evaluated by 99mTc-HMPAO labeling. Nucl Med Biol 35(1): 1175–1182.

Patki S, Kadam S, Chandra V & Bhonde R (2010) Human breast milk is a rich source of multipotent mesenchymal stem cells. Hum Cell 23(2): 35–40.

Petite H, Viateau V, Bensaid W, Meunier A, de Pollak C, Bourguignon M, Oudina K, Sedel L & Guillemin G (2000) Tissue-engineered bone regeneration. Nat Biotechnol 18(9): 959–963.

Pietila M, Lehenkari P, Kuvaja P, Kaakinen M, Kaul SC, Wadhwa R & Uemura T (2013) Mortalin antibody-conjugated quantum dot transfer from human mesenchymal stromal cells to breast cancer cells requires cell-cell interaction. Exp Cell Res 319(18): 2770–2780.

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S & Marshak DR (1999) Multilineage Potential of Adult Human Mesenchymal Stem Cells. Science 284(5411): 143–147.

Pokela M, Jantti V, Lepola P, Romsi P, Rimpilainen J, Kiviluoma K, Salomaki T, Vainionpaa V, Biancari F, Hirvonen J, Kaakinen T & Juvonen T (2003) EEG burst recovery is predictive of brain injury after experimental hypothermic circulatory arrest. Scand Cardiovasc J 37(3): 154–157.

Ponomaryov T, Peled A, Petit I, Taichman RS, Habler L, Sandbank J, Arenzana-Seisdedos F, Magerus A, Caruz A, Fujii N, Nagler A, Lahav M, Szyper-Kravitz M, Zipori D & Lapidot T (2000) Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function. J Clin Invest 106(11): 1331–1339.

Prockop DJ (1997) Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 276(5309): 71–74.

Qian H, Tryggvason K, Jacobsen SE & Ekblom M (2006) Contribution of alpha6 integrins to hematopoietic stem and progenitor cell homing to bone marrow and collaboration with alpha4 integrins. Blood 107(9): 3503–3510.

Page 126: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

124

Ramasamy R, Tong CK, Seow HF, Vidyadaran S & Dazzi F (2008) The immunosuppressive effects of human bone marrow-derived mesenchymal stem cells target T cell proliferation but not its effector function. Cell Immunol 251(2): 131–136.

Ranganath SH, Levy O, Inamdar MS & Karp JM (2012) Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell 10(3): 244–258.

Rao J, Lu L & Zhai Y (2014) T cells in organ ischemia reperfusion injury. Curr Opin Organ Transplant. 19(2):115–20

Reinders ME, de Fijter JW, Roelofs H, Bajema IM, de Vries DK, Schaapherder AF, Claas FH, van Miert PP, Roelen DL, van Kooten C, Fibbe WE & Rabelink TJ (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(2): 107–111.

Ren G, Su J, Zhang L, Zhao X, Ling W, L'huillie A, Zhang J, Lu Y, Roberts AI, Ji W, Zhang H, Rabson AB & Shi Y (2009) Species variation in the mechanisms of mesenchymal stem cell-mediated immunosuppression. Stem Cells 27(8): 1954–1962.

Ren G, Zhao X, Zhang L, Zhang J, L'Huillier A, Ling W, Roberts AI, Le AD, Shi S, Shao C & Shi Y (2010) Inflammatory cytokine-induced intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 in mesenchymal stem cells are critical for immunosuppression. J Immunol 184(5): 2321–2328.

Rerkasem K & Rothwell PM (2011) Carotid endarterectomy for symptomatic carotidstenosis. Cochrane Database Syst Rev. Apr 13;(4):CD001081

Ries C, Egea V, Karow M, Kolb H, Jochum M & Neth P (2007) MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood 109(9): 4055–4063.

Ringe J, Strassburg S, Neumann K, Endres M, Notter M, Burmester GR, Kaps C & Sittinger M (2007) Towards in situ tissue repair: human mesenchymal stem cells express chemokine receptors CXCR1, CXCR2 and CCR2, and migrate upon stimulation with CXCL8 but not CCL2. J Cell Biochem 101(1): 135–146.

Rojas M, Xu J, Woods CR, Mora AL, Spears W, Roman J & Brigham KL (2005) Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol 33(2): 145–152.

Rosado-de-Castro PH, Schmidt Fda R, Battistella V, Lopes de Souza SA, Gutfilen B, Goldenberg RC, Kasai-Brunswick TH, Vairo L, Silva RM, Wajnberg E, Alvarenga Americano do Brasil PE, Gasparetto EL, Maiolino A, Alves-Leon SV, Andre C, Mendez-Otero R, Rodriguez de Freitas G & Barbosa da Fonseca LM (2013) Biodistribution of bone marrow mononuclear cells after intra-arterial or intravenous transplantation in subacute stroke patients. Regen Med 8(2): 145–155.

Rose RA, Jiang H, Wang X, Helke S, Tsoporis JN, Gong N, Keating SC, Parker TG, Backx PH & Keating A (2008) Bone marrow-derived mesenchymal stromal cells express cardiac-specific markers, retain the stromal phenotype, and do not become functional cardiomyocytes in vitro. Stem Cells 26(11): 2884–2892.

Page 127: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

125

Rosova I, Dao M, Capoccia B, Link D & Nolta JA (2008) Hypoxic preconditioning results in increased motility and improved therapeutic potential of human mesenchymal stem cells. Stem Cells 26(8): 2173–2182.

Ruster B, Gottig S, Ludwig RJ, Bistrian R, Muller S, Seifried E, Gille J & Henschler R (2006) Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells. Blood 108(12): 3938–3944.

Sackstein R, Merzaban JS, Cain DW, Dagia NM, Spencer JA, Lin CP & Wohlgemuth R (2008) Ex vivo glycan engineering of CD44 programs human multipotent mesenchymal stromal cell trafficking to bone. Nat Med 14(2): 181–187.

Salem HK, Thiemermann C. Mesenchymal stromal cells: current understanding and clinical status. Stem Cells. 2010 Mar 31;28(3):585–96.

Sanchez-Ramos J, Song S, Cardozo-Pelaez F, Hazzi C, Stedeford T, Willing A, Freeman TB, Saporta S, Janssen W, Patel N, Cooper DR & Sanberg PR (2000) Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp Neurol 164(2): 247–256.

Sarkanen JR, Mannerstrom M, Vuorenpaa H, Uotila J, Ylikomi T & Heinonen T (2011) Intra-Laboratory Pre-Validation of a Human Cell Based in vitro Angiogenesis Assay for Testing Angiogenesis Modulators. Front Pharmacol 1: 147.

Sarkar D, Spencer JA, Phillips JA, Zhao W, Schafer S, Spelke DP, Mortensen LJ, Ruiz JP, Vemula PK, Sridharan R, Kumar S, Karnik R, Lin CP & Karp JM (2011) Engineered cell homing. Blood 118(25): e184–91.

Sarugaser R, Lickorish D, Baksh D, Hosseini MM & Davies JE (2005) Human umbilical cord perivascular (HUCPV) cells: a source of mesenchymal progenitors. Stem Cells 23(2): 220–229.

Schafer S, Calas AG, Vergouts M & Hermans E (2012) Immunomodulatory influence of bone marrow-derived mesenchymal stem cells on neuroinflammation in astrocyte cultures. J Neuroimmunol 249(1-2): 40–48.

Schrepfer S, Deuse T, Reichenspurner H, Fischbein MP, Robbins RC & Pelletier MP (2007) Stem cell transplantation: the lung barrier Transplant Proc 39(2): 573–576.

Schwarting S, Litwak S, Hao W, Bähr M, Weise J & Neumann H (2008) Hematopoietic Stem Cells Reduce Postischemic Inflammation and Ameliorate Ischemic Brain Injury. Stroke 39(10): 2867–2875.

Sharma A, Gokulchandran N, Chopra G, Kulkarni P, Lohia M, Badhe P & Jacob VC (2012) Administration of autologous bone marrow-derived mononuclear cells in children with incurable neurological disorders and injury is safe and improves their quality of life. Cell Transplant 21 Suppl 1: S79–90.

Shen LH, Li Y, Chen J, Zhang J, Vanguri P, Borneman J & Chopp M (2006) Intracarotid transplantation of bone marrow stromal cells increases axon-myelin remodeling after stroke. Neuroscience 137(2): 393–399.

Shih DT, Lee DC, Chen SC, Tsai RY, Huang CT, Tsai CC, Shen EY & Chiu WT (2005) Isolation and characterization of neurogenic mesenchymal stem cells in human scalp tissue. Stem Cells 23(7): 1012–1020.

Page 128: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

126

Shindo T, Matsumoto Y, Wang Q, Kawai N, Tamiya T & Nagao S (2006) Differences in the neuronal stem cells survival, neuronal differentiation and neurological improvement after transplantation of neural stem cells between mild and severe experimental traumatic brain injury. J Med Invest 53(1-2): 42–51.

Stemple DL & Anderson DJ (1992) Isolation of a stem cell for neurons and glia from the mammalian neural crest. Cell 71(6): 973–985.

Sundin M, Barrett AJ, Ringden O, Uzunel M, Lonnies H, Dackland AL, Christensson B & Blanc KL (2009) HSCT recipients have specific tolerance to MSC but not to the MSC donor. J Immunother 32(7): 755–764.

Tan J, Wu W, Xu X, Liao L, Zheng F, Messinger S, Sun X, Chen J, Yang S, Cai J, Gao X, Pileggi A & Ricordi C (2012) Induction therapy with autologous mesenchymal stem cells in living-related kidney transplants: a randomized controlled trial. JAMA 307(11): 1169–1177.

Tarkowski AK (1959) Experiments on the development of isolated blastomers of mouse eggs. Nature 184: 1286–1287.

Tatsumi K, Ohashi K, Matsubara Y, Kohori A, Ohno T, Kakidachi H, Horii A, Kanegae K, Utoh R, Iwata T & Okano T (2013) Tissue factor triggers procoagulation in transplanted mesenchymal stem cells leading to thromboembolism. Biochem Biophys Res Commun 431(2): 203–209.

Till JE & McCulloch EA (1961) A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat Res 14: 213–222.

Timmers L, Lim SK, Arslan F, Armstrong JS, Hoefer IE, Doevendans PA, Piek JJ, El Oakley RM, Choo A, Lee CN, Pasterkamp G & de Kleijn DP (2007) Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res 1(2): 129–137.

Tolar J, O'shaughnessy MJ, Panoskaltsis-Mortari A, McElmurry RT, Bell S, Riddle M, McIvor RS, Yant SR, Kay MA, Krause D, Verfaillie CM & Blazar BR (2006) Host factors that impact the biodistribution and persistence of multipotent adult progenitor cells. Blood 107(10): 4182–4188.

Toma C, Wagner WR, Bowry S, Schwartz A & Villanueva F (2009) Fate Of Culture-Expanded Mesenchymal Stem Cells in The Microvasculature. Circulation Research 104(3): 398–402.

van der Windt DJ, Bottino R, Casu A, Campanile N & Cooper DK (2007) Rapid loss of intraportally transplanted islets: an overview of pathophysiology and preventive strategies. Xenotransplantation 14(4): 288–297.

Vasconcelos-dos-Santos A, Rosado-de-Castro PH, Lopes de Souza SA, da Costa Silva J, Ramos AB, Rodriguez de Freitas G, Barbosa da Fonseca LM, Gutfilen B & Mendez-Otero R (2012) Intravenous and intra-arterial administration of bone marrow mononuclear cells after focal cerebral ischemia: Is there a difference in biodistribution and efficacy? Stem Cell Res 9(1): 1–8.

Page 129: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

127

Vilalta M, Degano IR, Bago J, Gould D, Santos M, Garcia-Arranz M, Ayats R, Fuster C, Chernajovsky Y, Garcia-Olmo D, Rubio N & Blanco J (2008) Biodistribution, long-term survival, and safety of human adipose tissue-derived mesenchymal stem cells transplanted in nude mice by high sensitivity non-invasive bioluminescence imaging. Stem Cells Dev 17(5): 993–1003.

von Bahr L, Batsis I, Moll G, Hagg M, Szakos A, Sundberg B, Uzunel M, Ringden O & Le Blanc K (2012) Analysis of tissues following mesenchymal stromal cell therapy in humans indicates limited long-term engraftment and no ectopic tissue formation. Stem Cells 30(7): 1575–1578.

Vorne M, Soini I, Lantto T & Paakkinen S (1989) Technetium-99m HM-PAO-labeled leukocytes in detection of inflammatory lesions: comparison with gallium-67 citrate J Nucl Med 30(8): 1332–1336.

Vulliet PR, Greeley M, Halloran SM, MacDonald KA & Kittleson MD (2004) Intra-coronary arterial injection of mesenchymal stromal cells and microinfarction in dogs. The Lancet 363(9411): 783–784.

Wang H, Cao F, De A, Cao Y, Contag C, Gambhir SS, Wu JC & Chen X (2009) Trafficking mesenchymal stem cell engraftment and differentiation in tumor-bearing mice by bioluminescence imaging. Stem Cells 27(7): 1548–1558.

Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, Fu YS, Lai MC & Chen CC (2004) Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord. Stem Cells 22(7): 1330–1337.

Wolf D, Reinhard A, Seckinger A, Katus HA, Kuecherer H & Hansen A (2009) Dose-dependent effects of intravenous allogeneic mesenchymal stem cells in the infarcted porcine heart. Stem Cells Dev 18(2): 321–329.

Woodbury D, Schwarz EJ, Prockop DJ & Black IB (2000) Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res 61(4): 364–370.

Yagi H, Soto-Gutierrez A, Kitagawa Y, Tilles AW, Tompkins RG & Yarmush ML (2010a) Bone marrow mesenchymal stromal cells attenuate organ injury induced by LPS and burn. Cell Transplant 19(6): 823–830.

Yagi H, Soto-Gutierrez A, Parekkadan B, Kitagawa Y, Tompkins RG, Kobayashi N & Yarmush ML (2010b) Mesenchymal stem cells: Mechanisms of immunomodulation and homing. Cell Transplant 19(6): 667–679.

Yang B, Strong R, Sharma S, Brenneman M, Mallikarjunarao K, Xi X, Grotta JC, Aronowski J & Savitz SI (2011) Therapeutic time window and dose response of autologous bone marrow mononuclear cells for ischemic stroke. J Neurosci Res 89(6): 833–839.

Zanardo G, Michielon P, Paccagnella A, Rosi P, Caló M, Salandin V, Da Ros A, Michieletto F & Simini G (1994) Acute renal failure in the patient undergoing cardiac operation. Prevalence, mortality rate, and main risk factors. J Thorac Cardiovasc Surg. 107(6):1489–95

Zhang ZG, Zhang L, Jiang Q, Zhang R, Davies K, Powers C, Bruggen N & Chopp M (2000) VEGF enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain. J Clin Invest 106(7): 829–838.

Page 130: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

128

Zhu H, Mitsuhashi N, Klein A, Barsky LW, Weinberg K, Barr ML, Demetriou A & Wu GD (2006) The role of the hyaluronan receptor CD44 in mesenchymal stem cell migration in the extracellular matrix. Stem Cells 24(4): 928–935.

Zimmerlin L, Donnenberg AD, Rubin JP, Basse P, Landreneau RJ & Donnenberg VS (2011) Regenerative therapy and cancer: in vitro and in vivo studies of the interaction between adipose-derived stem cells and breast cancer cells from clinical isolates. Tissue Eng Part A 17(1-2): 93–106.

Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P & Hedrick MH (2002) Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 13(12): 4279–4295.

Page 131: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

129

Original publications

I Mäkelä T, Takalo R, Arvola O, Haapanen H, Yannopoulos F, Blanco R, Ahvenjärvi L, Lehtonen S, Kiviluoma K, Kerkelä E, Nystedt J, Juvonen T & Lehenkari P. (2014) Safety and Biodistribution Study of Bone Marrow Derived Mesenchymal Stromal Cells and Mononuclear Cells and the Impact of the Administration Route in an Intact Porcine Model. Manuscript

II Mäkelä T, Yannopoulos F, Alestalo K, Mäkelä J, Lepola P, Anttila V, Lehtonen S, Kiviluoma K, Takalo R, Juvonen T & Lehenkari P. (2013) Intra-Arterial Bone Marrow Mononuclear Cell Distribution after Experimental Global Brain Ischemia Scand Cardiovasc J. 47(2):114-20.

III Kerkelä E, Hakkarainen T, Mäkelä T, Kilpinen L, Lehtonen S, Ritamo I, Pernu R, Pietilä M, Takalo R, Nikkilä J, Tikkanen J, Juvonen T, Laitinen S, Valmu L, Lehenkari P & Nystedt J. (2013) Transient Proteolytic Modification of Mesenchymal Stromal Cells Decreases Lung Entrapment in vivo. Stem Cells Translational Medicine. 2(7):510-20.

Reprinted with permission from Informa Healthcare (II) and AlphaMed Press (III)

Original publications are not included in the elecronic version of the dissertation.

Page 132: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

130

Page 133: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

A C T A U N I V E R S I T A T I S O U L U E N S I S

Book orders:Granum: Virtual book storehttp://granum.uta.fi/granum/

S E R I E S D M E D I C A

1256. Karhu, Jaana (2014) Severe community- acquired pneumonia – studies on imaging,etiology, treatment, and outcome among intensive care patients

1257. Lahti, Anniina (2014) Epidemiological study on trends and characteristics ofsuicide among children and adolescents in Finland

1258. Nyyssönen, Virva (2014) Transvaginal mesh-augmented procedures in gynecology :outcomes after female urinary incontinence and pelvic organ prolapse surgery

1259. Kummu, Outi (2014) Humoral immune response to carbamyl-epitopes inatherosclerosis

1260. Jokinen, Elina (2014) Targeted therapy sensitivity and resistance in solidmalignancies

1261. Amegah, Adeladza Kofi (2014) Household fuel and garbage combustion, streetvending activities and adverse pregnancy outcomes : evidence from urban Ghana

1262. Roisko, Riikka (2014) Parental Communication Deviance as a risk factor forthought disorders and schizophrenia spectrum disorders in offspring : The FinnishAdoptive Family Study

1263. Åström, Pirjo (2014) Regulatory mechanisms mediating matrix metalloproteinase-8 effects in oral tissue repair and tongue cancer

1264. Haikola, Britta (2014) Oral health among Finns aged 60 years and older :edentulousness, fixed prostheses, dental infections detected from radiographsand their associating factors

1265. Manninen, Anna-Leena (2014) Clinical applications of radiophotoluminescence(RPL) dosimetry in evaluation of patient radiation exposure in radiology :Determination of absorbed and effective dose

1266. Kuusisto, Sanna (2014) Effects of heavy alcohol intake on lipoproteins,adiponectin and cardiovascular risk

1267. Kiviniemi, Marjo (2014) Mortality, disability, psychiatric treatment and medicationin first-onset schizophrenia in Finland : the register linkage study

1268. Kallio, Merja (2014) Neurally adjusted ventilatory assist in pediatric intensive care

1269. Väyrynen, Juha (2014) Immune cell infiltration and inflammatory biomarkers incolorectal cancer

1270. Silvola, Anna-Sofia (2014) Effect of treatment of severe malocclusion and relatedfactors on oral health-related quality of life

Page 134: OULU 2014 D 1271 ACTA UNIVERSITATIS OULUENSIS …jultika.oulu.fi/files/isbn9789526206547.pdf · 2015. 12. 16. · A very special thanks goes to Seija Seljänperä for acting as the

ABCDEFG

UNIVERSITY OF OULU P .O. B 00 F I -90014 UNIVERSITY OF OULU FINLAND

A C T A U N I V E R S I T A T I S O U L U E N S I S

S E R I E S E D I T O R S

SCIENTIAE RERUM NATURALIUM

HUMANIORA

TECHNICA

MEDICA

SCIENTIAE RERUM SOCIALIUM

SCRIPTA ACADEMICA

OECONOMICA

EDITOR IN CHIEF

PUBLICATIONS EDITOR

Professor Esa Hohtola

University Lecturer Santeri Palviainen

Postdoctoral research fellow Sanna Taskila

Professor Olli Vuolteenaho

University Lecturer Veli-Matti Ulvinen

Director Sinikka Eskelinen

Professor Jari Juga

Professor Olli Vuolteenaho

Publications Editor Kirsti Nurkkala

ISBN 978-952-62-0653-0 (Paperback)ISBN 978-952-62-0654-7 (PDF)ISSN 0355-3221 (Print)ISSN 1796-2234 (Online)

U N I V E R S I TAT I S O U L U E N S I S

MEDICA

ACTAD

D 1271

ACTA

Tuomas M

äkelä

OULU 2014

D 1271

Tuomas Mäkelä

SYSTEMIC TRANSPLANTATION OF BONE MARROW STROMAL CELLSAN EXPERIMENTAL ANIMAL STUDY OF BIODISTRIBUTION AND TISSUE TARGETING

UNIVERSITY OF OULU GRADUATE SCHOOL;UNIVERSITY OF OULU,FACULTY OF MEDICINE,INSTITUTE OF CLINICAL MEDICINE,DEPARTMENT OF SURGERY,DEPARTMENT OF ANAESTHESIOLOGY;INSTITUTE OF BIOMEDICINE,DEPARTMENT OF ANATOMY AND CELL BIOLOGY;INSTITUTE OF DIAGNOSTICS,DEPARTMENT OF DIAGNOSTIC RADIOLOGY;CLINICAL RESEARCH CENTER