1
|
Lange M, Granados A, VijayKumar S, Bragantini J, Ancheta S, Kim YJ, Santhosh S, Borja M, Kobayashi H, McGeever E, Solak AC, Yang B, Zhao X, Liu Y, Detweiler AM, Paul S, Theodoro I, Mekonen H, Charlton C, Lao T, Banks R, Xiao S, Jacobo A, Balla K, Awayan K, D'Souza S, Haase R, Dizeux A, Pourquie O, Gómez-Sjöberg R, Huber G, Serra M, Neff N, Pisco AO, Royer LA. A multimodal zebrafish developmental atlas reveals the state-transition dynamics of late-vertebrate pluripotent axial progenitors. Cell 2024; 187:6742-6759.e17. [PMID: 39454574 DOI: 10.1016/j.cell.2024.09.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 05/02/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024]
Abstract
Elucidating organismal developmental processes requires a comprehensive understanding of cellular lineages in the spatial, temporal, and molecular domains. In this study, we introduce Zebrahub, a dynamic atlas of zebrafish embryonic development that integrates single-cell sequencing time course data with lineage reconstructions facilitated by light-sheet microscopy. This atlas offers high-resolution and in-depth molecular insights into zebrafish development, achieved through the sequencing of individual embryos across ten developmental stages, complemented by reconstructions of cellular trajectories. Zebrahub also incorporates an interactive tool to navigate the complex cellular flows and lineages derived from light-sheet microscopy data, enabling in silico fate-mapping experiments. To demonstrate the versatility of our multimodal resource, we utilize Zebrahub to provide fresh insights into the pluripotency of neuro-mesodermal progenitors (NMPs) and the origins of a joint kidney-hemangioblast progenitor population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Bin Yang
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Xiang Zhao
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Yang Liu
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Sheryl Paul
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | | | | | - Tiger Lao
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Sheng Xiao
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Keir Balla
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Kyle Awayan
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Robert Haase
- Cluster of Excellence "Physics of Life," TU Dresden, Dresden, Germany
| | - Alexandre Dizeux
- Institute of Physics for Medicine Paris, ESPCI Paris-PSL, Paris, France
| | | | | | - Greg Huber
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Mattia Serra
- University of California, San Diego, San Diego, CA, USA
| | - Norma Neff
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | | |
Collapse
|
2
|
Rosner M, Horer S, Feichtinger M, Hengstschläger M. Multipotent fetal stem cells in reproductive biology research. Stem Cell Res Ther 2023; 14:157. [PMID: 37287077 DOI: 10.1186/s13287-023-03379-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Abstract
Due to the limited accessibility of the in vivo situation, the scarcity of the human tissue, legal constraints, and ethical considerations, the underlying molecular mechanisms of disorders, such as preeclampsia, the pathological consequences of fetomaternal microchimerism, or infertility, are still not fully understood. And although substantial progress has already been made, the therapeutic strategies for reproductive system diseases are still facing limitations. In the recent years, it became more and more evident that stem cells are powerful tools for basic research in human reproduction and stem cell-based approaches moved into the center of endeavors to establish new clinical concepts. Multipotent fetal stem cells derived from the amniotic fluid, amniotic membrane, chorion leave, Wharton´s jelly, or placenta came to the fore because they are easy to acquire, are not associated with ethical concerns or covered by strict legal restrictions, and can be banked for autologous utilization later in life. Compared to adult stem cells, they exhibit a significantly higher differentiation potential and are much easier to propagate in vitro. Compared to pluripotent stem cells, they harbor less mutations, are not tumorigenic, and exhibit low immunogenicity. Studies on multipotent fetal stem cells can be invaluable to gain knowledge on the development of dysfunctional fetal cell types, to characterize the fetal stem cells migrating into the body of a pregnant woman in the context of fetomaternal microchimerism, and to obtain a more comprehensive picture of germ cell development in the course of in vitro differentiation experiments. The in vivo transplantation of fetal stem cells or their paracrine factors can mediate therapeutic effects in preeclampsia and can restore reproductive organ functions. Together with the use of fetal stem cell-derived gametes, such strategies could once help individuals, who do not develop functional gametes, to conceive genetically related children. Although there is still a long way to go, these developments regarding the usage of multipotent fetal stem cells in the clinic should continuously be accompanied by a wide and detailed ethical discussion.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Stefanie Horer
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | | | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| |
Collapse
|
3
|
A novel type of mesenchymal stem cells derived from bovine metanephric mesenchyme. Tissue Cell 2022; 79:101970. [DOI: 10.1016/j.tice.2022.101970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/15/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
|
4
|
Mattonet K, Riemslagh FW, Guenther S, Prummel KD, Kesavan G, Hans S, Ebersberger I, Brand M, Burger A, Reischauer S, Mosimann C, Stainier DYR. Endothelial versus pronephron fate decision is modulated by the transcription factors Cloche/Npas4l, Tal1, and Lmo2. SCIENCE ADVANCES 2022; 8:eabn2082. [PMID: 36044573 PMCID: PMC9432843 DOI: 10.1126/sciadv.abn2082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/11/2022] [Indexed: 05/17/2023]
Abstract
Endothelial specification is a key event during embryogenesis; however, when, and how, endothelial cells separate from other lineages is poorly understood. In zebrafish, Npas4l is indispensable for endothelial specification by inducing the expression of the transcription factor genes etsrp, tal1, and lmo2. We generated a knock-in reporter in zebrafish npas4l to visualize endothelial progenitors and their derivatives in wild-type and mutant embryos. Unexpectedly, we find that in npas4l mutants, npas4l reporter-expressing cells contribute to the pronephron tubules. Single-cell transcriptomics and live imaging of the early lateral plate mesoderm in wild-type embryos indeed reveals coexpression of endothelial and pronephron markers, a finding confirmed by creERT2-based lineage tracing. Increased contribution of npas4l reporter-expressing cells to pronephron tubules is also observed in tal1 and lmo2 mutants and is reversed in npas4l mutants injected with tal1 mRNA. Together, these data reveal that Npas4l/Tal1/Lmo2 regulate the fate decision between the endothelial and pronephron lineages.
Collapse
Affiliation(s)
- Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- DZHK (German Center for Cardiovascular Research), partner site, 43, D-61231 Bad Nauheim
- CPI (Cardio Pulmonary Institute), partner site, 43, D-61231 Bad Nauheim
- DZL (German Center for Lung Research), partner site, 43, D-61231 Bad Nauheim
| | - Fréderike W. Riemslagh
- Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Stefan Guenther
- DZHK (German Center for Cardiovascular Research), partner site, 43, D-61231 Bad Nauheim
- CPI (Cardio Pulmonary Institute), partner site, 43, D-61231 Bad Nauheim
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Karin D. Prummel
- Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Gokul Kesavan
- Center for Regenerative Therapies at TU Dresden (CRTD); Dresden, Germany
| | - Stefan Hans
- Center for Regenerative Therapies at TU Dresden (CRTD); Dresden, Germany
| | - Ingo Ebersberger
- Goethe University Frankfurt am Main, Institute of Cell Biology and Neuroscience, Frankfurt 60438, Germany
- Senckenberg Biodiversity and Climate Research Center (S-BIKF), Frankfurt 60325, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Frankfurt 60325, Germany
| | - Michael Brand
- Center for Regenerative Therapies at TU Dresden (CRTD); Dresden, Germany
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- CPI (Cardio Pulmonary Institute), partner site, 43, D-61231 Bad Nauheim
| | - Christian Mosimann
- Section of Developmental Biology, Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- DZHK (German Center for Cardiovascular Research), partner site, 43, D-61231 Bad Nauheim
- CPI (Cardio Pulmonary Institute), partner site, 43, D-61231 Bad Nauheim
- DZL (German Center for Lung Research), partner site, 43, D-61231 Bad Nauheim
| |
Collapse
|
5
|
Therapeutic Mesenchymal Stromal Cells for Immunotherapy and for Gene and Drug Delivery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 16:204-224. [PMID: 32071924 PMCID: PMC7012781 DOI: 10.1016/j.omtm.2020.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs) possess several fairly unique properties that, when combined, make them ideally suited for cellular-based immunotherapy and as vehicles for gene and drug delivery for a wide range of diseases and disorders. Key among these are: (1) their relative ease of isolation from a variety of tissues; (2) the ability to be expanded in culture without a loss of functionality, a property that varies to some degree with tissue source; (3) they are relatively immune-inert, perhaps obviating the need for precise donor/recipient matching; (4) they possess potent immunomodulatory functions that can be tailored by so-called licensing in vitro and in vivo; (5) the efficiency with which they can be modified with viral-based vectors; and (6) their almost uncanny ability to selectively home to damaged tissues, tumors, and metastases following systemic administration. In this review, we summarize the latest research in the immunological properties of MSCs, their use as immunomodulatory/anti-inflammatory agents, methods for licensing MSCs to customize their immunological profile, and their use as vehicles for transferring both therapeutic genes in genetic disease and drugs and genes designed to destroy tumor cells.
Collapse
|
6
|
Boruczkowski D, Zdolińska-Malinowska I. A Retrospective Analysis of Safety and Efficacy of Wharton's Jelly Stem Cell Administration in Children with Spina Bifida. Stem Cell Rev Rep 2019; 15:717-729. [PMID: 31222411 DOI: 10.1007/s12015-019-09902-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aim of this paper was to describe the outcome of therapeutic administration of mesenchymal stem cells (MSC) obtained from Wharton's jelly (WJ-MSCs) in paediatric patients with spina bifida (SB) during a medical therapeutic experiment. We retrospectively analysed the records of twenty-eight patients aged 1-18 years (median age 4 years) recruited in daily clinical practice. Each patient received 0.9-5.0 × 106 WJ-MSCs/kg (median 2.6 × 106 WJ-MSCs/kg) administered in 1-5 injections as an experimental treatment for SB (allogenic administration). All the patients were examined by the same neurologist (study investigator, SI) on the day of each infusion. Based on the neurological examination, the SI used a six-point Likert scale to assess the quality of life and self-service of each patient. Twenty-six follow-up observations after MSC administration were analysed retrospectively. In addition, the assessments of the parents and other healthcare professionals were obtained for 5 patients and compared with the SI's assessment. Twenty-one of 26 patients (81%) experienced some improvement in their health status. Twenty-one (81%) patients experienced increased quality of life (median 2.0) and 10 patients (38%) achieved a slight increase in their self-service level (median 1). Improvement was achieved in 12 out of 17 areas. Five were significant in low-power sign test: muscle tension, muscle strength, gross motor development, micturition/defecation control, and cognitive functions. Adverse events were mild and temporary. Age, body mass, single dose or poor response after the first administration were not significant predictors of later response to treatment in contrast to the total cell dose per one kg in the whole treatment course. WJ-MSC administration is a safe and effective procedure that improves motor functions, micturition/defecation control, and cognitive functions, and improves the quality of life in children with SB.
Collapse
Affiliation(s)
- Dariusz Boruczkowski
- Medical Department, Polski Bank Komórek Macierzystych S.A. (FamiCord Group), Jana Pawła II 29, 00-867, Warsaw, Poland.
| | - Izabela Zdolińska-Malinowska
- Medical Department, Polski Bank Komórek Macierzystych S.A. (FamiCord Group), Jana Pawła II 29, 00-867, Warsaw, Poland
| |
Collapse
|
7
|
Almeida-Porada G, Rodman C, Kuhlman B, Brudvik E, Moon J, George S, Guida P, Sajuthi SP, Langefeld CD, Walker SJ, Wilson PF, Porada CD. Exposure of the Bone Marrow Microenvironment to Simulated Solar and Galactic Cosmic Radiation Induces Biological Bystander Effects on Human Hematopoiesis. Stem Cells Dev 2018; 27:1237-1256. [PMID: 29698131 DOI: 10.1089/scd.2018.0005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The stem cell compartment of the hematopoietic system constitutes one of the most radiosensitive tissues of the body and leukemias represent one of the most frequent radiogenic cancers with short latency periods. As such, leukemias may pose a particular threat to astronauts during prolonged space missions. Control of hematopoiesis is tightly governed by a specialized bone marrow (BM) microenvironment/niche. As such, any environmental insult that damages cells of this niche would be expected to produce pronounced effects on the types and functionality of hematopoietic/immune cells generated. We recently reported that direct exposure of human hematopoietic stem cells (HSC) to simulated solar energetic particle (SEP) and galactic cosmic ray (GCR) radiation dramatically altered the differentiative potential of these cells, and that simulated GCR exposures can directly induce DNA damage and mutations within human HSC, which led to leukemic transformation when these cells repopulated murine recipients. In this study, we performed the first in-depth examination to define changes that occur in mesenchymal stem cells present in the human BM niche following exposure to accelerated protons and iron ions and assess the impact these changes have upon human hematopoiesis. Our data provide compelling evidence that simulated SEP/GCR exposures can also contribute to defective hematopoiesis/immunity through so-called "biological bystander effects" by damaging the stromal cells that comprise the human marrow microenvironment, thereby altering their ability to support normal hematopoiesis.
Collapse
Affiliation(s)
- Graça Almeida-Porada
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Christopher Rodman
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Bradford Kuhlman
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Egil Brudvik
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - John Moon
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Sunil George
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Peter Guida
- 2 Biological, Environmental, and Climate Sciences Department, Brookhaven National Laboratory , Upton, New York
| | - Satria P Sajuthi
- 3 Division of Public Health Sciences, Department of Biostatistical Sciences, Center for Public Health Genomics , Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Carl D Langefeld
- 3 Division of Public Health Sciences, Department of Biostatistical Sciences, Center for Public Health Genomics , Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Stephen J Walker
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Paul F Wilson
- 4 Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center , Sacramento, California
| | - Christopher D Porada
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| |
Collapse
|
8
|
Bettini S, Franceschini V, Astolfi L, Simoni E, Mazzanti B, Martini A, Revoltella RP. Human mesenchymal stromal cell therapy for damaged cochlea repair in nod-scid mice deafened with kanamycin. Cytotherapy 2017; 20:189-203. [PMID: 29246648 DOI: 10.1016/j.jcyt.2017.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/05/2017] [Accepted: 11/03/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Kanamycin, mainly used in the treatment of drug-resistant-tuberculosis, is known to cause irreversible hearing loss. Using the xeno-transplant model, we compared both in vitro and in vivo characteristics of human mesenchymal stromal cells (MSCs) derived from adult tissues, bone marrow (BM-MSCs) and adipose tissue (ADSCs). These tissues were selected for their availability, in vitro multipotency and regenerative potential in vivo in kanamycin-deafened nod-scid mice. METHODS MSCs were isolated from informed donors and expanded ex vivo. We evaluated their proliferation capacity in vitro using the hexosaminidase assay, the phenotypic profile using flow-cytometry of a panel of surface antigens, the osteogenic potential using alkaline phosphatase activity and the adipogenic potential using oil-red-O staining. MSCs were intravenously injected in deafened mice and cochleae, liver, spleen and kidney were sampled 7 and 30 days after transplantation. The dissected organs were analyzed using lectin histochemistry, immunohistochemistry, polymerase chain reaction (PCR) and dual color fluorescence in situ hybridization (DC-FISH). RESULTS MSCs showed similar in vitro characteristics, but ADSCs appeared to be more efficient after prolonged expansion. Both cell types engrafted in the cochlea of damaged mice, inducing regeneration of the damaged sensory structures. Several hybrid cells were detected in engrafted tissues. DISCUSSION BM-MSCs and ADSCs showed in vitro characteristics suitable for tissue regeneration and fused with resident cells in engrafted tissues. The data suggest that paracrine effect is the prevalent mechanism inducing tissue recovery. Overall, BM-MSCs and ADSCs appear to be valuable tools in regenerative medicine for hearing loss recovery.
Collapse
Affiliation(s)
- Simone Bettini
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Valeria Franceschini
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy; Foundation Onlus 'Staminali e Vita', Padua, Italy.
| | - Laura Astolfi
- Foundation Onlus 'Staminali e Vita', Padua, Italy; Bioacoustics Research Laboratory, Department of Neurosciences, University of Padua, Padua, Italy
| | - Edi Simoni
- Bioacoustics Research Laboratory, Department of Neurosciences, University of Padua, Padua, Italy
| | - Benedetta Mazzanti
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Alessandro Martini
- Foundation Onlus 'Staminali e Vita', Padua, Italy; Bioacoustics Research Laboratory, Department of Neurosciences, University of Padua, Padua, Italy
| | - Roberto P Revoltella
- Foundation Onlus 'Staminali e Vita', Padua, Italy; Institute for Chemical, Physical Processes, Centro Nazionale delle Ricerche (C.N.R.), Pisa, Italy
| |
Collapse
|
9
|
Abstract
BACKGROUND In utero transplantation (IUT) of hematopoietic stem cells has the potential to treat a large number of hematologic and metabolic diseases amenable to partial replacement of the hematopoietic system. METHODS A review of the literature was conducted that focused on the clinical and experimental experience with IUT and, in this context, the development of the hematopoietic and immune systems. RESULTS Successful application of IUT has been limited to the treatment of various types of immunodeficiencies that affect lymphocyte development and function. Other congenital defects such as the thalassemias have not resulted in clinically significant engraftment. Recent efforts at understanding and overcoming the barriers to engraftment in the fetus have focused on providing a selective advantage to donor stem cells and fostering immune tolerance toward the donor cells. The critical cellular components of the graft that promote engraftment and tolerance induction are being evaluated in animal models. Improvements in engraftment have resulted from the inclusion of T cells and/or dendritic cells in the graft, as well as a strategy of combined prenatal and postnatal transplantation. CONCLUSIONS The advantages, necessity, and benefits of early treatment will continue to encourage development of IUT as a means to treat hematopoietic and other types of birth defects.
Collapse
Affiliation(s)
- Marcus O Muench
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA.
| | | |
Collapse
|
10
|
Almeida-Porada G, Atala A, Porada CD. In utero stem cell transplantation and gene therapy: rationale, history, and recent advances toward clinical application. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 5:16020. [PMID: 27069953 PMCID: PMC4813605 DOI: 10.1038/mtm.2016.20] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/29/2016] [Accepted: 01/29/2016] [Indexed: 12/11/2022]
Abstract
Recent advances in high-throughput molecular testing have made it possible to diagnose most genetic disorders relatively early in gestation with minimal risk to the fetus. These advances should soon allow widespread prenatal screening for the majority of human genetic diseases, opening the door to the possibility of treatment/correction prior to birth. In addition to the obvious psychological and financial benefits of curing a disease in utero, and thereby enabling the birth of a healthy infant, there are multiple biological advantages unique to fetal development, which provide compelling rationale for performing potentially curative treatments, such as stem cell transplantation or gene therapy, prior to birth. Herein, we briefly review the fields of in utero transplantation (IUTx) and in utero gene therapy and discuss the biological hurdles that have thus far restricted success of IUTx to patients with immunodeficiencies. We then highlight several recent experimental breakthroughs in immunology, hematopoietic/marrow ontogeny, and in utero cell delivery, which have collectively provided means of overcoming these barriers, thus setting the stage for clinical application of these highly promising therapies in the near future.
Collapse
Affiliation(s)
- Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina, USA
| | - Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine , Winston Salem, North Carolina, USA
| |
Collapse
|
11
|
Girlovanu M, Susman S, Soritau O, Rus-Ciuca D, Melincovici C, Constantin AM, Mihu CM. Stem cells - biological update and cell therapy progress. ACTA ACUST UNITED AC 2015; 88:265-71. [PMID: 26609255 PMCID: PMC4632881 DOI: 10.15386/cjmed-483] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/12/2015] [Indexed: 12/17/2022]
Abstract
In recent years, the advances in stem cell research have suggested that the human body may have a higher plasticity than it was originally expected. Until now, four categories of stem cells were isolated and cultured in vivo: embryonic stem cells, fetal stem cells, adult stem cells and induced pluripotent stem cells (hiPSCs). Although multiple studies were published, several issues concerning the stem cells are still debated, such as: the molecular mechanisms of differentiation, the methods to prevent teratoma formation or the ethical and religious issues regarding especially the embryonic stem cell research. The direct differentiation of stem cells into specialized cells: cardiac myocytes, neural cells, pancreatic islets cells, may represent an option in treating incurable diseases such as: neurodegenerative diseases, type I diabetes, hematologic or cardiac diseases. Nevertheless, stem cell-based therapies, based on stem cell transplantation, remain mainly at the experimental stages and their major limitation is the development of teratoma and cancer after transplantation. The induced pluripotent stem cells (hiPSCs) represent a prime candidate for future cell therapy research because of their significant self-renewal and differentiation potential and the lack of ethical issues. This article presents an overview of the biological advances in the study of stem cells and the current progress made in the field of regenerative medicine.
Collapse
Affiliation(s)
- Mihai Girlovanu
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sergiu Susman
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Olga Soritau
- Research Department, Prof. Dr. I. Chiricuta Oncology Institute, Cluj-Napoca, Romania
| | - Dan Rus-Ciuca
- Department of Pathology, Karlstad Central Hospital, Sweden
| | - Carmen Melincovici
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anne-Marie Constantin
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Carmen Mihaela Mihu
- Morphological Sciences Department 1, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
12
|
Placenta as a Source of Stem Cells for Regenerative Medicine. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0070-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
13
|
Porada CD, Rodman C, Ignacio G, Atala A, Almeida-Porada G. Hemophilia A: an ideal disease to correct in utero. Front Pharmacol 2014; 5:276. [PMID: 25566073 PMCID: PMC4263089 DOI: 10.3389/fphar.2014.00276] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/27/2014] [Indexed: 01/13/2023] Open
Abstract
Hemophilia A (HA) is the most frequent inheritable defect of the coagulation proteins. The current standard of care for patients with HA is prophylactic factor infusion, which is comprised of regular (2-3 times per week) intravenous infusions of recombinant or plasma-derived FVIII to maintain hemostasis. While this treatment has greatly increased the quality of life and lengthened the life expectancy for many HA patients, its high cost, the need for lifelong infusions, and the fact that it is unavailable to roughly 75% of the world's HA patients make this type of treatment far from ideal. In addition, this lifesaving therapy suffers from a high risk of treatment failure due to immune response to the infused FVIII. There is thus a need for novel treatments, such as those using stem cells and/or gene therapy, which have the potential to mediate long-term correction or permanent cure following a single intervention. In the present review, we discuss the clinical feasibility and unique advantages that an in utero approach to treating HA could offer, placing special emphasis on a new sheep model of HA we have developed and on the use of mesenchymal stromal cells (MSC) as cellular vehicles for delivering the FVIII gene.
Collapse
Affiliation(s)
| | | | | | | | - Graça Almeida-Porada
- Regenerative Medicine, Wake Forest Institute for Regenerative MedicineWinston-Salem, NC, USA
| |
Collapse
|
14
|
Valim V, Amorin B, Pezzi A, Aparecida Lima da Silva M, Paula Alegretti A, Silla L. Optimization of the Cultivation of Donor Mesenchymal Stromal Cells for Clinical Use in Cellular Therapy. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/cellbio.2014.31003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
15
|
Harari-Steinberg O, Metsuyanim S, Omer D, Gnatek Y, Gershon R, Pri-Chen S, Ozdemir DD, Lerenthal Y, Noiman T, Ben-Hur H, Vaknin Z, Schneider DF, Aronow BJ, Goldstein RS, Hohenstein P, Dekel B. Identification of human nephron progenitors capable of generation of kidney structures and functional repair of chronic renal disease. EMBO Mol Med 2013; 5:1556-68. [PMID: 23996934 PMCID: PMC3799579 DOI: 10.1002/emmm.201201584] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 07/29/2013] [Accepted: 07/31/2013] [Indexed: 12/14/2022] Open
Abstract
Identification of tissue-specific renal stem/progenitor cells with nephrogenic potential is a critical step in developing cell-based therapies for renal disease. In the human kidney, stem/progenitor cells are induced into the nephrogenic pathway to form nephrons until the 34 week of gestation, and no equivalent cell types can be traced in the adult kidney. Human nephron progenitor cells (hNPCs) have yet to be isolated. Here we show that growth of human foetal kidneys in serum-free defined conditions and prospective isolation of NCAM1(+) cells selects for nephron lineage that includes the SIX2-positive cap mesenchyme cells identifying a mitotically active population with in vitro clonogenic and stem/progenitor properties. After transplantation in the chick embryo, these cells-but not differentiated counterparts-efficiently formed various nephron tubule types. hNPCs engrafted and integrated in diseased murine kidneys and treatment of renal failure in the 5/6 nephrectomy kidney injury model had beneficial effects on renal function halting disease progression. These findings constitute the first definition of an intrinsic nephron precursor population, with major potential for cell-based therapeutic strategies and modelling of kidney disease.
Collapse
Affiliation(s)
- Orit Harari-Steinberg
- The Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Center for Regenerative Medicine, Sheba Medical CenterRamat-Gan, Israel
| | - Sally Metsuyanim
- The Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Center for Regenerative Medicine, Sheba Medical CenterRamat-Gan, Israel
- Mina and Everard Goodman Faculty of Life Sciences, Bar-IlanUniversityRamat-Gan, Israel
| | - Dorit Omer
- The Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Center for Regenerative Medicine, Sheba Medical CenterRamat-Gan, Israel
- Sackler School of Medicine, Tel Aviv UniversityTel Aviv, Israel
| | - Yehudit Gnatek
- The Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Center for Regenerative Medicine, Sheba Medical CenterRamat-Gan, Israel
| | - Rotem Gershon
- The Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Center for Regenerative Medicine, Sheba Medical CenterRamat-Gan, Israel
- Sackler School of Medicine, Tel Aviv UniversityTel Aviv, Israel
| | - Sara Pri-Chen
- The Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Center for Regenerative Medicine, Sheba Medical CenterRamat-Gan, Israel
| | - Derya D Ozdemir
- The Roslin Institute, University of Edinburgh, Easter Bush CampusMidlothian, UK
| | - Yaniv Lerenthal
- Cancer Research Center, Sheba Medical CenterRamat-Gan, Israel
| | - Tzahi Noiman
- Mina and Everard Goodman Faculty of Life Sciences, Bar-IlanUniversityRamat-Gan, Israel
| | - Herzel Ben-Hur
- L.E.M. Laboratory of Early DetectionNes Ziona, Israel
- Department of Obstet and Gynecology, Assaf HarofehTzrifin, Israel
| | - Zvi Vaknin
- Department of Obstet and Gynecology, Assaf HarofehTzrifin, Israel
| | | | - Bruce J Aronow
- Division of Molecular and Developmental Biology, Department of Pediatrics, University of Cincinnati, Childrens Hospital Medical CenterCincinnati, OH, USA
| | - Ronald S Goldstein
- Mina and Everard Goodman Faculty of Life Sciences, Bar-IlanUniversityRamat-Gan, Israel
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Easter Bush CampusMidlothian, UK
| | - Benjamin Dekel
- The Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Center for Regenerative Medicine, Sheba Medical CenterRamat-Gan, Israel
- Sackler School of Medicine, Tel Aviv UniversityTel Aviv, Israel
- Division of Pediatric Nephrology, Edmond& Lily Safra Children's Hospital, Sheba Medical CenterRamat-Gan, Israel
| |
Collapse
|
16
|
Colletti E, El Shabrawy D, Soland M, Yamagami T, Mokhtari S, Osborne C, Schlauch K, Zanjani ED, Porada CD, Almeida-Porada G. EphB2 isolates a human marrow stromal cell subpopulation with enhanced ability to contribute to the resident intestinal cellular pool. FASEB J 2013; 27:2111-21. [PMID: 23413357 DOI: 10.1096/fj.12-205054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
To identify human bone marrow stromal cell (BMSC) subsets with enhanced ability to engraft/contribute to the resident intestinal cellular pool, we transplanted clonally derived BMSCs into fetal sheep. Analysis at 75 d post-transplantation showed 2 of the 6 clones engrafting the intestine at 4- to 5-fold higher levels (5.03±0.089 and 5.04±0.15%, respectively) than the other clones (P<0.01), correlating with the percentage of donor-derived Musashi-1(+) (12.01-14.17 vs. 1.2-3.8%; P<0.01) or leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5)(+) cells within the intestinal stem cell (ISC) region. Phenotypic and transcriptome analysis determined that the clones with enhanced intestinal contribution expressed high levels of Ephrin type B receptor 2 (EphB2). Intestinal explants demonstrated proliferation of the engrafted cells and ability to generate crypt-like structures in vitro still expressing EphB2. Additional transplants based on BMSC EphB2 expression demonstrated that, at 7 d post-transplant, the EphB2(high) BMSCs engrafted in the ISC region at levels of 2.1 ± 0.2%, while control EphB2(low) BMSCs engrafted at 0.3 ± 0.1% (P<0.01). Therefore we identified a marker for isolating and culturing an expandable subpopulation of BMSCs with enhanced intestinal homing and contribution to the ISC region.
Collapse
Affiliation(s)
- Evan Colletti
- Department of Animal Biotechnology, University of Nevada, Reno, Nevada, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Porada CD, Almeida-Porada G. Treatment of Hemophilia A in Utero and Postnatally using Sheep as a Model for Cell and Gene Delivery. ACTA ACUST UNITED AC 2013; S1. [PMID: 23264887 DOI: 10.4172/2157-7412.s1-011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hemophilia A represents the most common inheritable deficiency of the coagulation proteins. Current state-of- the-art treatment consists of frequent prophylactic infusions of plasma-derived or recombinant FVIII protein to maintain hemostasis, and has greatly increased life expectancy and quality of life for many hemophilia A patients. This treatment approach is, however, far from ideal, due to the need for lifelong intravenous infusions, the high treatment cost, and the fact that it is unavailable to a large percentage of the world's hemophiliacs. There is thus a need for novel treatments that can promise long-term or permanent correction. In contrast to existing protein based therapeutics, gene therapy offers to provide a permanent cure following few, or even a single, treatment. In the present paper, we review ongoing work towards this end, focusing on studies we have performed in a large animal model. Some of the key topics covered in this review include the unique opportunities sheep offer as a model system, the re-establishment and clinical and molecular characterization of a line of sheep with severe hemophilia A, the advantages and feasibility of treating a disease like hemophilia A in utero, and the use of Mesenchymal Stem Cells (MSC) as cellular delivery vehicles for the FVIII gene. The review finishes with a brief discussion of our recent success correcting ovine hemophilia A with a postnatal transplant with gene-modified MSC, and the limitations of this approach that remain to be overcome.
Collapse
|
18
|
Yang N, Xu L, Lin P, Cui J. Uric acid promotes neuronal differentiation of human placenta-derived mesenchymal stem cells in a time- and concentration-dependent manner. Neural Regen Res 2012; 7:756-60. [PMID: 25737698 PMCID: PMC4345657 DOI: 10.3969/j.issn.1673-5374.2012.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 02/22/2012] [Indexed: 12/23/2022] Open
Abstract
Uric acid is an important, naturally occurring serum antioxidant. The present study investigates the use of uric acid for promoting proliferation and neuronal differentiation of mesenchymal stem cells derived from human placenta tissue. Human placenta-derived mesenchymal stem cells were pre-induced in the presence of either 0, 0.2, 0.4 or 0.8 mM uric acid in combination with 1 mM β-mercaptoethanol for 24 hours, followed by exposure to identical uric acid concentrations and 5 mM β-mercaptoethanol for 6 and 10 hours. Cells developed a neuronal-like morphology, with formation of interconnected process extensions, typical of neural cells. Immunocytochemistry and immunofluorescence staining showed neuron specific enolase positive cells were present in each group except the control group. A greater number of neuron specific enolase positive cells were observed in 0.8 mM uric acid in combination with 5 mM β-mercaptoethanol at 10 hours. After 24 hours of induction, Nissl bodies were detected in the cytoplasm of all differentiated cell groups except the control group and Nissl body numbers were greatest in human placenta-derived mesenchymal stem cells grown in the presence of 0.8 mM uric acid and 5 mM β-mercaptoethanol. These results suggest uric acid accelerates differentiation of human placenta-derived mesenchymal stem cells into neuronal-like cells in a time- and concentration-dependent manner.
Collapse
Affiliation(s)
- Nailong Yang
- Department of Endocrinology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Lili Xu
- Department of Endocrinology, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| | - Peng Lin
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan 250002, Shandong Province, China
| | - Jing Cui
- VIP Department, Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, Shandong Province, China
| |
Collapse
|
19
|
Zhang ZY, Teoh SH, Hui JHP, Fisk NM, Choolani M, Chan JKY. The potential of human fetal mesenchymal stem cells for off-the-shelf bone tissue engineering application. Biomaterials 2012; 33:2656-72. [PMID: 22217806 DOI: 10.1016/j.biomaterials.2011.12.025] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 12/13/2011] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have become one of the most promising cell sources for bone tissue engineering (BTE) applications. In this review, we first highlight recent progress in the understanding of MSC biology, their in vivo niche, multi-faceted contribution to fracture healing and bone re-modelling, and their role in BTE. A literature review from clinicaltrials.gov and Pubmed on clinical usage of MSC for both orthopedic and non-orthopedic indications suggests that translational use of MSC for BTE indications is likely to bear fruit in the ensuing decade. Last, we disscuss the profound influence of ontological and antomical origins of MSC on their proliferation and osteogenesis and demonstrated human fetal MSC (hfMSC) as a superior cellular candidate for off-the-shelf BTE applications. This relates to their superior proliferation capacity, more robust osteogenic potential and lower immunogenecity, as compared to MSC from perinatal and postnatal sources. Furthermore, we discuss our experience in developing a hfMSC based BTE strategy with the integrated use of bioreactor-based dynamic priming within macroporous scaffolds, now ready for evaluation in clinical trials. In conclusion, hfMSC is likely the most promising cell source for allogeneic based BTE application, with proven advantages compared to other MSC based ones.
Collapse
Affiliation(s)
- Zhi-Yong Zhang
- Mechanical Engineering, Faculty of Engineering, National University of Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
20
|
Wenceslau CV, Miglino MA, Martins DS, Ambrósio CE, Lizier NF, Pignatari GC, Kerkis I. Mesenchymal Progenitor Cells from Canine Fetal Tissues: Yolk Sac, Liver, and Bone Marrow. Tissue Eng Part A 2011; 17:2165-76. [DOI: 10.1089/ten.tea.2010.0678] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Cristiane Valverde Wenceslau
- Departments of Surgery and Pathology, University of São Paulo, São Paulo, Brazil
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC), Ribeirão Preto, Brazil
| | - Maria Angélica Miglino
- Departments of Surgery and Pathology, University of São Paulo, São Paulo, Brazil
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC), Ribeirão Preto, Brazil
| | - Daniele Santos Martins
- Faculty of Animal Sciences and Food Engineering, University of São Paulo, São Paulo, Brazil
| | - Carlos Eduardo Ambrósio
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC), Ribeirão Preto, Brazil
- Faculty of Animal Sciences and Food Engineering, University of São Paulo, São Paulo, Brazil
| | | | | | - Irina Kerkis
- Faculty of Animal Sciences and Food Engineering, University of São Paulo, São Paulo, Brazil
- Laboratory of Genetics, Butantan Institute, São Paulo, Brazil
| |
Collapse
|
21
|
Porada CD, Almeida-Porada G. Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery. Adv Drug Deliv Rev 2010; 62:1156-66. [PMID: 20828588 DOI: 10.1016/j.addr.2010.08.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Revised: 08/26/2010] [Accepted: 08/27/2010] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) possess a set of several fairly unique properties which make them ideally suited both for cellular therapies/regenerative medicine, and as vehicles for gene and drug delivery. These include: 1) relative ease of isolation; 2) the ability to differentiate into a wide variety of seemingly functional cell types of both mesenchymal and non-mesenchymal origin; 3) the ability to be extensively expanded in culture without a loss of differentiative capacity; 4) they are not only hypoimmunogenic, but they produce immunosuppression upon transplantation; 5) their pronounced anti-inflammatory properties; and 6) their ability to home to damaged tissues, tumors, and metastases following in vivo administration. In this review, we summarize the latest research in the use of mesenchymal stem cells in regenerative medicine, as immunomodulatory/anti-inflammatory agents, and as vehicles for transferring both therapeutic genes in genetic disease and genes designed to destroy malignant cells.
Collapse
|
22
|
The umbilical cord matrix is a better source of mesenchymal stem cells (MSC) than the umbilical cord blood. Cell Biol Int 2010; 34:693-701. [PMID: 20187873 DOI: 10.1042/cbi20090414] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Many studies have drawn attention to the emerging role of MSC (mesenchymal stem cells) as a promising population supporting new clinical concepts in cellular therapy. However, the sources from which these cells can be isolated are still under discussion. Whereas BM (bone marrow) is presented as the main source of MSC, despite the invasive procedure related to this source, the possibility of isolating sufficient numbers of these cells from UCB (umbilical cord blood) remains controversial. Here, we present the results of experiments aimed at isolating MSC from UCB, BM and UCM (umbilical cord matrix) using different methods of isolation and various culture media that summarize the main procedures and criteria reported in the literature. Whereas isolation of MSC were successful from BM (10:10) and (UCM) (8:8), only one cord blood sample (1:15) gave rise to MSC using various culture media [DMEM (Dulbecco's modified Eagle's medium) +5% platelet lysate, DMEM+10% FBS (fetal bovine serum), DMEM+10% human UCB serum, MSCGM] and different isolation methods [plastic adherence of total MNC (mononuclear cells), CD3+/CD19+/CD14+/CD38+-depleted MNC and CD133+- or LNGFR+-enriched MNC]. MSC from UCM and BM were able to differentiate into adipocytes, osteocytes and hepatocytes. The expansion potential was highest for MSC from UCM. The two cell populations had CD90+/CD73+/CD105+ phenotype with the additional expression of SSEA4 and LNGFR for BM MSC. These results clearly exclude UCB from the list of MSC sources for clinical use and propose instead UCM as a rich, non-invasive and abundant source of MSC.
Collapse
|
23
|
Abdulrazzak H, Moschidou D, Jones G, Guillot PV. Biological characteristics of stem cells from foetal, cord blood and extraembryonic tissues. J R Soc Interface 2010; 7 Suppl 6:S689-706. [PMID: 20739312 DOI: 10.1098/rsif.2010.0347.focus] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Foetal stem cells (FSCs) can be isolated during gestation from many different tissues such as blood, liver and bone marrow as well as from a variety of extraembryonic tissues such as amniotic fluid and placenta. Strong evidence suggests that these cells differ on many biological aspects such as growth kinetics, morphology, immunophenotype, differentiation potential and engraftment capacity in vivo. Despite these differences, FSCs appear to be more primitive and have greater multi-potentiality than their adult counterparts. For example, foetal blood haemopoietic stem cells proliferate more rapidly than those found in cord blood or adult bone marrow. These features have led to FSCs being investigated for pre- and post-natal cell therapy and regenerative medicine applications. The cells have been used in pre-clinical studies to treat a wide range of diseases such as skeletal dysplasia, diaphragmatic hernia and respiratory failure, white matter damage, renal pathologies as well as cancers. Their intermediate state between adult and embryonic stem cells also makes them an ideal candidate for reprogramming to the pluripotent status.
Collapse
Affiliation(s)
- Hassan Abdulrazzak
- Institute of Reproductive and Developmental Biology, Imperial College London, London W12 0NN, UK
| | | | | | | |
Collapse
|
24
|
Lee ESM, Bou-Gharios G, Seppanen E, Khosrotehrani K, Fisk NM. Fetal stem cell microchimerism: natural-born healers or killers? Mol Hum Reprod 2010; 16:869-78. [PMID: 20663958 DOI: 10.1093/molehr/gaq067] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Eddy S M Lee
- University of Queensland Centre for Clinical Research, University of Queensland, Herston campus, Brisbane 4029, Australia
| | | | | | | | | |
Collapse
|
25
|
Almeida-Porada G, Zanjani ED, Porada CD. Bone marrow stem cells and liver regeneration. Exp Hematol 2010; 38:574-80. [PMID: 20417684 PMCID: PMC2882990 DOI: 10.1016/j.exphem.2010.04.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 04/07/2010] [Accepted: 04/13/2010] [Indexed: 12/11/2022]
Abstract
Development of new approaches to treat patients with hepatic diseases that can eliminate the need for liver transplantation is imperative. Use of cell therapy as a means of repopulating the liver has several advantages over whole-organ transplantation because it would be less invasive, less immunogenic, and would allow the use, in some instances, of autologous-derived cells. Stem/progenitor cells that would be ideal for liver repopulation would need to have characteristics such as availability and ease of isolation, the ability to be expanded in vitro, ensuring adequate numbers of cells, susceptibility to modification by viral vector transduction/genetic recombination, to correct any underlying genetic defects, and the ability of restoring liver function following transplantation. Bone marrow-derived stem cells, such as hematopoietic, mesenchymal and endothelial progenitor cells possess some or most of these characteristics, making them ideal candidates for liver regenerative therapies. Here, we will summarize the ability of each of these stem cell populations to give rise to functional hepatic elements that could mediate repair in patients with liver damage/disease.
Collapse
Affiliation(s)
- Graça Almeida-Porada
- Department of Animal Biotechnology, University of Nevada, Reno, Reno, NV 89557-0104, USA.
| | | | | |
Collapse
|
26
|
Franceschini V, Bettini S, Pifferi S, Rosellini A, Menini A, Saccardi R, Ognio E, Jeffery R, Poulsom R, Revoltella RP. Human cord blood CD133+ stem cells transplanted to nod-scid mice provide conditions for regeneration of olfactory neuroepithelium after permanent damage induced by dichlobenil. Stem Cells 2009; 27:825-35. [PMID: 19350683 DOI: 10.1002/stem.11] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The herbicide dichlobenil selectively causes necrosis of the dorsomedial part of olfactory neuroepithelium (NE) with permanent damage to the underlying mucosa, whereas the lateral part of the olfactory region and the nasal respiratory mucosa remain undamaged. We investigated here whether human umbilical cord blood CD133(+) stem cells (HSC) injected intravenously to nod-scid mice pretreated with dichlobenil may engraft the olfactory mucosa and contribute to the regeneration of the damaged NE. We tested HLA-DQalpha1 DNA and three human microsatellites (Combined DNA Index System) as indicators of engrafted cells, finding polymerase chain reaction evidence of chimaerism in various tissues of the host, including the olfactory mucosa and bulb, at 7 and 31 days following HSC transplantation. Histology, immunohistochemistry, and lectin staining revealed the morphological recovery of the dorsomedial region of the NE in dichlobenil-treated mice that received HSC, contrasting with the lack of regeneration in similarly injured areas as these remained damaged in control nontransplanted mice. FISH analysis, to detect human genomic sequences from different chromosomes, confirmed persistent engraftment of the regenerating olfactory area with chimeric cells. Electro-olfactograms in response to odorants, to test the functionality of the olfactory NE, confirmed the functional damage of the dorsomedial area in dichlobenil-treated mice and the functional recovery of the same area in transplanted mice. These findings support the concept that transplanted HSC migrating to the damaged olfactory area provide conditions facilitating the recovery from olfactory receptor cell loss.
Collapse
Affiliation(s)
- Valeria Franceschini
- Department of Experimental Evolutionary Biology, University of Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Mizokami T, Hisha H, Okazaki S, Takaki T, Wang XL, Song CY, Li Q, Kato J, Hosaka N, Inaba M, Kanzaki H, Ikehara S. Preferential expansion of human umbilical cord blood-derived CD34-positive cells on major histocompatibility complex-matched amnion-derived mesenchymal stem cells. Haematologica 2009; 94:618-28. [PMID: 19336739 DOI: 10.3324/haematol.2008.004705] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND We previously found in a murine hematopoietic system that hematopoietic stem cells show high differentiation and proliferation capacity on bone marrow-derived mesenchymal stem cells/stromal cells (microenvironment) with "self" major histocompatibility complex (MHC). DESIGN AND METHODS We examined whether amnion-derived adherent cells have the characteristics of mesenchymal stem cells, and whether these adherent cells can support the proliferation of umbilical cord blood-derived lineage-negative and CD34-positive cells (Lin(-)CD34(+) cells) obtained from the same fetus to a greater extent than those derived from other fetuses. RESULTS Culture-expanded amnion-derived adherent cells expressed mesenchymal stem cell markers and HLA-ABC molecules and could differentiate into osteoblasts, adipocytes and chondrocyte-like cells, indicating that the cells have the characteristics of mesenchymal stem cells. The Lin(-)CD34(+) cells purified from the frozen umbilical cord blood were strongly positive for HLA-ABC, and contained a large number of hematopoietic stem cells. When the Lin(-)CD34(+) cells were cultured on the autologous (MHC-matched) or MHC-mismatched amnion-derived adherent cells in short-term assays (hematopoietic stem cell-proliferation) and long-term culture-initiating cell assays, greater expansion of the Lin(-)CD34(+) cells was observed in the MHC-matched combination than in MHC-mismatched combinations. The concentration of granulocyte-macrophage colony-stimulating factor in the culture supernatants of the long-term culture-initiating cell assays was significantly higher in the MHC-matched combination than in MHC-mismatched combinations. CONCLUSIONS IT is likely that a MHC restriction exists between hematopoietic stem cells and mesenchymal stem cells/stromal cells in the human hematopoietic system and that granulocute-macropage colony-stimulating factor contributes to some extent to the preferential hematopoiesis-supporting ability of the MHC-matched amnion-derived adherent cells.
Collapse
Affiliation(s)
- Tomomi Mizokami
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi City, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gucciardo L, Lories R, Ochsenbein-Kölble N, Done' E, Zwijsen A, Deprest J. Fetal mesenchymal stem cells: isolation, properties and potential use in perinatology and regenerative medicine. BJOG 2009; 116:166-72. [PMID: 19076948 DOI: 10.1111/j.1471-0528.2008.02005.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The fetus is a source of nonembryonic stem cells (SC), with potential applications in perinatal medicine. Cells derived from the placenta, membranes, amniotic fluid or fetal tissues are higher in number, expansion potential and differentiation abilities compared with SC from adult tissues. Although some obstacles keep SC biology at distance from clinical application, the feasibility of using (homologous) SC for tissue engineering for the fetus with a congenital birth defect has been demonstrated. Also, other pathologies may benefit from SC technology.
Collapse
Affiliation(s)
- L Gucciardo
- Department of Obstetrics and Gynecology, University Hospital Gasthuisberg, Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
29
|
Colletti EJ, Airey JA, Liu W, Simmons PJ, Zanjani ED, Porada CD, Almeida-Porada G. Generation of tissue-specific cells from MSC does not require fusion or donor-to-host mitochondrial/membrane transfer. Stem Cell Res 2008; 2:125-38. [PMID: 19383418 DOI: 10.1016/j.scr.2008.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Revised: 08/27/2008] [Accepted: 08/27/2008] [Indexed: 12/17/2022] Open
Abstract
Human mesenchymal stem cells (MSC) hold great promise for cellular replacement therapies. Despite their contributing to phenotypically distinct cells in multiple tissues, controversy remains regarding whether the phenotype switch results from a true differentiation process. Here, we studied the events occurring during the first 120 h after human MSC transplantation into a large animal model. We demonstrate that MSC, shortly after engrafting different tissues, undergo proliferation and rapidly initiate the differentiative process, changing their phenotype into tissue-specific cells. Thus, the final level of tissue-specific cell contribution is not determined solely by the initial level of engraftment of the MSC within that organ, but rather by the proliferative capability of the ensuing tissue-specific cells into which the MSC rapidly differentiate. Furthermore, we show that true differentiation, and not cell fusion or transfer of mitochondria or membrane-derived vesicles between transplanted and resident cells, is the primary mechanism contributing to the change of phenotype of MSC upon transplantation.
Collapse
Affiliation(s)
- Evan J Colletti
- Department of Animal Biotechnology, University of Nevada at Reno, Reno, NV 89557, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Application potential of human fetal stem/progenitor cells in cell therapy. Bull Exp Biol Med 2008; 145:114-21. [DOI: 10.1007/s10517-008-0031-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
31
|
Revoltella RP, Papini S, Rosellini A, Michelini M, Franceschini V, Ciorba A, Bertolaso L, Magosso S, Hatzopoulos S, Lorito G, Giordano P, Simoni E, Ognio E, Cilli M, Saccardi R, Urbani S, Jeffery R, Poulsom R, Martini A. Cochlear Repair by Transplantation of Human Cord Blood CD133+ Cells to Nod-Scid Mice Made Deaf with Kanamycin and Noise. Cell Transplant 2008; 17:665-78. [DOI: 10.3727/096368908786092685] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We investigated the fate of human cord blood CD133+ hematopoietic stem cells (HSC) transplanted intravenously (IV) into irradiated nodscid mice previously made deaf by ototoxic treatment with kanamycin and/or intense noise, to verify whether HSC engraft the cochlea and contribute to inner ear restoration, in vivo. We tested the presence of HLA.DQα1 by PCR, used for traceability of engrafted cells, finding evidence that HSC migrated to various host tissues, including the organ of Corti (OC). By histology, antibody and lectin-staining analysis, we confirmed that HSC IV transplantation in mice previously damaged by ototoxic agents correlated with the repair process and stimulation ex novo of morphological recovery in the inner ear, while the cochlea of control oto-injured, nontransplanted mice remained seriously damaged. Dual color FISH analysis also provided evidence of positive engraftment in the inner ear and in various mouse tissues, also revealing small numbers of heterokaryons, probably derived from fusion of donor with endogenous cells, for up to 2 months following transplantation. These observations offer the first evidence that transplanted human HSC migrating to the inner ear of oto-injured mice may provide conditions for the resumption of deafened cochlea, emerging as a potential strategy for inner ear rehabilitation.
Collapse
Affiliation(s)
- Roberto P. Revoltella
- Foundation onlus “Staminali e Vita”, Institute of Biomedical Technologies, C.N.R., 56127 Pisa, Italy
| | - Sandra Papini
- Foundation onlus “Staminali e Vita”, Institute of Biomedical Technologies, C.N.R., 56127 Pisa, Italy
| | - Alfredo Rosellini
- Foundation onlus “Staminali e Vita”, Institute of Biomedical Technologies, C.N.R., 56127 Pisa, Italy
| | - Monica Michelini
- Foundation onlus “Staminali e Vita”, Institute of Biomedical Technologies, C.N.R., 56127 Pisa, Italy
| | - Valeria Franceschini
- Department of Evolution and Experimental Biology, University of Bologna, 40126 Bologna, Italy
| | - Andrea Ciorba
- Audiology Unit, Bioacoustic Center and National Institute of Neuroscience, University Hospital S. Anna, University of Ferrara, 44100 Ferrara, Italy
| | - Lucia Bertolaso
- Audiology Unit, Bioacoustic Center and National Institute of Neuroscience, University Hospital S. Anna, University of Ferrara, 44100 Ferrara, Italy
| | - Sara Magosso
- Audiology Unit, Bioacoustic Center and National Institute of Neuroscience, University Hospital S. Anna, University of Ferrara, 44100 Ferrara, Italy
| | - Stavros Hatzopoulos
- Audiology Unit, Bioacoustic Center and National Institute of Neuroscience, University Hospital S. Anna, University of Ferrara, 44100 Ferrara, Italy
| | - Guiscardo Lorito
- Audiology Unit, Bioacoustic Center and National Institute of Neuroscience, University Hospital S. Anna, University of Ferrara, 44100 Ferrara, Italy
| | - Pietro Giordano
- Audiology Unit, Bioacoustic Center and National Institute of Neuroscience, University Hospital S. Anna, University of Ferrara, 44100 Ferrara, Italy
| | - Edi Simoni
- Audiology Unit, Bioacoustic Center and National Institute of Neuroscience, University Hospital S. Anna, University of Ferrara, 44100 Ferrara, Italy
| | - Emanuela Ognio
- Animal Model Facility, National Institute for Cancer Research (IST), 16132 Genua, Italy
| | - Michele Cilli
- Animal Model Facility, National Institute for Cancer Research (IST), 16132 Genua, Italy
| | - Riccardo Saccardi
- Bone Marrow Transplantation Center, Department of Haematology, University Hospital, 50134 Florence, Italy
| | - Serena Urbani
- Bone Marrow Transplantation Center, Department of Haematology, University Hospital, 50134 Florence, Italy
| | - Rosemary Jeffery
- Histopathology Unit, Cancer Research UK, London Research Institute, London WC2A 3PX, UK
| | - Richard Poulsom
- Histopathology Unit, Cancer Research UK, London Research Institute, London WC2A 3PX, UK
| | - Alessandro Martini
- Histopathology Unit, Cancer Research UK, London Research Institute, London WC2A 3PX, UK
| |
Collapse
|
32
|
Gubhaju L, Laslett A, Bertram JF, Zulli A, Black MJ. Immunohistochemical localisation of TRA-1-60, TRA-1-81, GCTM-2 and podocalyxin in the developing baboon kidney. Histochem Cell Biol 2008; 129:651-7. [PMID: 18265998 DOI: 10.1007/s00418-008-0398-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2008] [Indexed: 10/22/2022]
Abstract
The baboon is an ideal animal model to study human kidney development. The aim of the current study was to use immunohistochemistry to localise the antigens TRA-1-60, TRA-1-81, GCTM-2 and podocalyxin in the developing baboon kidney where nephrogenesis was still on-going and in kidneys where nephrogenesis was complete. Fixed kidney sections from baboons delivered at 125, 140, 175 and 185 days gestation (term = 185 days) were immuno-labelled with antibodies directed against TRA-1-60, TRA-1-81, GCTM-2 and podocalyxin. In kidneys with on-going nephrogenesis (125 and 140 days gestation), TRA-1-60, TRA-1-81 and GCTM-2 were specifically localised to the apical plasma membrane of the epithelium of the ureteric ampullae and the collecting ducts, while podocalyxin immunostaining was not detected. In kidneys where nephrogenesis was complete (175 and 185 days gestation) localisation of these markers was again very specifically localised to the collecting ducts. In conclusion, although further experimentation is required to confirm the identity of the specific cell types marked by these antibodies, this study provides new insight into the distribution of commonly utilised stem cell antibodies in the developing baboon kidney.
Collapse
Affiliation(s)
- Lina Gubhaju
- Department of Anatomy and Developmental Biology, Monash University, PO Box 13C, Clayton, VIC 3800, Australia.
| | | | | | | | | |
Collapse
|
33
|
Chamberlain J, Yamagami T, Colletti E, Theise ND, Desai J, Frias A, Pixley J, Zanjani ED, Porada CD, Almeida-Porada G. Efficient generation of human hepatocytes by the intrahepatic delivery of clonal human mesenchymal stem cells in fetal sheep. Hepatology 2007; 46:1935-45. [PMID: 17705296 DOI: 10.1002/hep.21899] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Alternative methods to whole liver transplantation require a suitable cell that can be expanded to obtain sufficient numbers required for successful transplantation while maintaining the ability to differentiate into hepatocytes. Mesenchymal stem cells (MSCs) possess several advantageous characteristics for cell-based therapy and have been shown to be able to differentiate into hepatocytes. Thus, we investigated whether the intrahepatic delivery of human MSCs is a safe and effective method for generating human hepatocytes and whether the route of administration influences the levels of donor-derived hepatocytes and their pattern of distribution throughout the parenchyma of the recipient's liver. Human clonally derived MSCs were transplanted by an intraperitoneal (n = 6) or intrahepatic (n = 6) route into preimmune fetal sheep. The animals were analyzed 56-70 days after transplantation by immunohistochemistry, enzyme-linked immunosorbent assay, and flow cytometry. The intrahepatic injection of human MSCs was safe and resulted in more efficient generation of hepatocytes (12.5% +/- 3.5% versus 2.6% +/- 0.4%). The animals that received an intrahepatic injection exhibited a widespread distribution of hepatocytes throughout the liver parenchyma, whereas an intraperitoneal injection resulted in a preferential periportal distribution of human hepatocytes that produced higher amounts of albumin. Furthermore, hepatocytes were generated from MSCs without the need to first migrate/lodge to the bone marrow and give rise to hematopoietic cells. CONCLUSION Our studies provide evidence that MSCs are a valuable source of cells for liver repair and regeneration and that, by the alteration of the site of injection, the generation of hepatocytes occurs in different hepatic zones, suggesting that a combined transplantation approach may be necessary to successfully repopulate the liver with these cells.
Collapse
Affiliation(s)
- Jason Chamberlain
- Department of Animal Biotechnology, University of Nevada at Reno, Reno, NV 89557-0104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Almeida-Porada G, Porada C, Gupta N, Torabi A, Thain D, Zanjani ED. The human-sheep chimeras as a model for human stem cell mobilization and evaluation of hematopoietic grafts' potential. Exp Hematol 2007; 35:1594-600. [PMID: 17889724 PMCID: PMC2048750 DOI: 10.1016/j.exphem.2007.07.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Revised: 06/06/2007] [Accepted: 07/13/2007] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To investigate whether the sheep xenograft model of human hematopoiesis can be used to mimic mobilization of human hematopoietic stem cells in vivo. MATERIAL AND METHODS Sheep transplanted with 3.6 x 10(6) CD34+ from human adult bone marrow were mobilized 1.5 years posttransplantation with human granulocyte colony-stimulating factor for 5 days. At day 3 and 4 of mobilization, human cells were harvested from peripheral blood (PB) and bone marrow (BM) and were injected into secondary sheep recipients (n = 6) and these animals were analyzed for the presence of human cells in their BM and PB, starting at 3.5 months posttransplantation. RESULTS Maximum mobilization of human cells in PB occurred at day 3, with a 21-fold increase in total numbers of human cells, and a recovery of 5.5 x 10(4)/mL CD34+. In the BM, maximal numbers of human cells were achieved at day 4, with a 6.3-fold increase and a recovery of 1.5 x 10(4)/mL CD34+ cells. PB and BM mobilized human cells were then transplanted into new sheep recipients, and analysis at 3.5 months posttransplantation demonstrated that levels of human cell engraftment in BM of the group transplanted with mobilized PB were significantly lower than those transplanted with BM cells (0.6% +/- 0.1% vs 8.0% +/- 1.8%). Furthermore, in sheep transplanted with mobilized PB, the levels of human cells in circulation remained 2.5-fold higher than the levels of human cells found in their BM. CONCLUSION Mobilization of human cells in the sheep model parallels human PB and BM hematopoietic stem cells (HSC) mobilization in healthy human donors in their ability to engraft, differentiate, and repopulate secondary hosts. Thus, this model can become a useful tool to study mobilization regimens, mechanisms, and quality of products obtained.
Collapse
Affiliation(s)
- Graça Almeida-Porada
- Department of Animal Biotechnology University of Nevada, Reno, NV 89557-0104, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Chen B, Shi J, Zheng J, Chen Y, Wang K, Yang Q, Chen X, Yang Z, Zhou X, Zhu Y, Chu J, Liu A, Sheng HZ. Differentiation of liver cells from human primordial germ cell-derived progenitors. Differentiation 2007; 75:350-9. [PMID: 17286597 DOI: 10.1111/j.1432-0436.2006.00151.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In previous studies, progenitor embryoid body-derived (EBD) cells have been derived from human embryonic germ cells. These cells express lineage markers of three primary germ layers, although their potential to produce true fetal cells of various types has yet to be tested. To this end, we have transplanted EBD cells into the fetal sheep liver. We show that these cells respond appropriately to environmental cues and give rise to hepatocytes and well-structured bile ducts. These results suggest that EBD cells are relatively uncommitted early progenitors capable of effective incorporation and differentiation in vivo. The ability to generate functional liver cells makes EBD cells potentially useful for cell therapy.
Collapse
Affiliation(s)
- Bin Chen
- Center for Developmental Biology, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, 1665 Kong Jiang Road, Shanghai 200092, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Dekel B, Zangi L, Shezen E, Reich-Zeliger S, Eventov-Friedman S, Katchman H, Jacob-Hirsch J, Amariglio N, Rechavi G, Margalit R, Reisner Y. Isolation and characterization of nontubular sca-1+lin- multipotent stem/progenitor cells from adult mouse kidney. J Am Soc Nephrol 2006; 17:3300-14. [PMID: 17093069 DOI: 10.1681/asn.2005020195] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Tissue engineering and cell therapy approaches aim to take advantage of the repopulating ability and plasticity of multipotent stem cells to regenerate lost or diseased tissue. Recently, stage-specific embryonic kidney progenitor tissue was used to regenerate nephrons. Through fluorescence-activated cell sorting, microarray analysis, in vitro differentiation assays, mixed lymphocyte reaction, and a model of ischemic kidney injury, this study sought to identify and characterize multipotent organ stem/progenitor cells in the adult kidney. Herein is reported the existence of nontubular cells that express stem cell antigen-1 (Sca-1). This population of small cells includes a CD45-negative fraction that lacks hematopoietic stem cell and lineage markers and resides in the renal interstitial space. In addition, these cells are enriched for beta1-integrin, are cytokeratin negative, and show minimal expression of surface markers that typically are found on bone marrow-derived mesenchymal stem cells. Global gene profiling reveals enrichment for many genes downstream of developmental signaling molecules and self-renewal pathways, such as TGF-beta/bone morphogenic protein, Wnt, or fibroblast growth factor, as well as for those that are involved in specification of mesodermal lineages (myocyte enhancer factor 2A, YY1-associated factor 2, and filamin-beta). In vitro, they are plastic adherent and slowly proliferating and result in inhibition of alloreactive CD8(+) T cells, indicative of an immune-privileged behavior. Furthermore, clonal-derived lines can be differentiated into myogenic, osteogenic, adipogenic, and neural lineages. Finally, when injected directly into the renal parenchyma, shortly after ischemic/reperfusion injury, renal Sca-1(+)Lin(-) cells, derived from ROSA26 reporter mice, adopt a tubular phenotype and potentially could contribute to kidney repair. These data define a unique phenotype for adult kidney-derived cells, which have potential as stem cells and may contribute to the regeneration of injured kidneys.
Collapse
Affiliation(s)
- Benjamin Dekel
- Weizmann Institute of Science, Department of Immunology, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zeng F, Chen MJ, Baldwin DA, Gong ZJ, Yan JB, Qian H, Wang J, Jiang X, Ren ZR, Sun D, Huang SZ. Multiorgan engraftment and differentiation of human cord blood CD34+ Lin- cells in goats assessed by gene expression profiling. Proc Natl Acad Sci U S A 2006; 103:7801-6. [PMID: 16682618 PMCID: PMC1472525 DOI: 10.1073/pnas.0602646103] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
To investigate multitissue engraftment of human primitive hematopoietic cells and their differentiation in goats, human CD34+ Lin- cord blood cells transduced with a GFP vector were transplanted into fetal goats at 45-55 days of gestation. GFP+ cells were detected in hematopoietic and nonhematopoietic organs including blood, bone marrow, spleen, liver, kidney, muscle, lung, and heart of the recipient goats (1.2-36% of all cells examined). We identified human beta2 microglobulin-positive cells in multiple tissues. GFP+ cells sorted from the perfused liver of a transplant goat showed human insulin-like growth factor 1 gene sequences, indicating that the engrafted GFP+ cells were of human origin. A substantial fraction of cells engrafted in goat livers expressed the human hepatocyte-specific antigen, proliferating cell nuclear antigen, albumin, hepatocyte nuclear factor, and GFP. DNA content analysis showed no evidence for cellular fusion. Long-term engraftment of GFP+ cells could be detected in the blood of goats for up to 2 yr. Microarray analysis indicated that human genes from a variety of functional categories were expressed in chimeric livers and blood. The human/goat xenotransplant model provides a unique system to study the kinetics of hematopoietic stem cell engraftment, gene expression, and possible stem cell plasticity under noninjured conditions.
Collapse
Affiliation(s)
- Fanyi Zeng
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, 24/1400 West Beijing Road, Shanghai 200040, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Teague WJ, Rowan-Hull AM, Jayanthi NVG, Johnson PRV. The competency of foregut mesenchyme in islet mesenchyme-to-epithelial transition during embryonic development. J Pediatr Surg 2006; 41:347-51. [PMID: 16481249 DOI: 10.1016/j.jpedsurg.2005.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND/PURPOSE Potential for curative stem-cell treatments of juvenile-onset diabetes has focussed research into pancreatic islet development. Islets were previously thought to originate solely from embryonic pancreatic epithelium, but we have demonstrated that islets can originate from mesenchyme, that is, islet mesenchyme-to-epithelial transition. The aim of this study was to establish the competence of foregut mesenchyme during mesenchymal islet development. METHODS Embryonic chick pancreatic epithelium of gestational stage Hamburger-Hamilton (HH) 22 (J Morphol. 1951;88:49-92) was combined with quail stomach mesenchyme of increasing gestation (stage HH22 [n = 6], HH26 [n = 6], HH28 [n = 4], or HH31 [n = 6]). Recombinants were cultured and analysed by immunocytochemistry for coexpression of insulin and quail-specific antigen to determine the embryonic origin of islets. RESULTS Recombinants constructed using stage HH22 mesenchyme yielded 34 islets, of which 35% were mesenchymal. However, when recombinants were constructed using stage HH26 mesenchyme, 24% of 25 islets were mesenchymal. When using mesenchyme, 13% of 15 islets were mesenchymal. All islets (n = 35) in recombinants constructed using stage HH31 mesenchyme were epithelial derived. Islet mesenchyme-to-epithelial transition diminished significantly with increasing mesenchymal gestational stage (P = .002). CONCLUSIONS These data show foregut mesenchyme is competent to form islets between stages HH22 and HH28. Developmental competence of foregut mesenchyme in islet mesenchyme-to-epithelial transition diminishes as gestation increases. This may have important implications for identifying stem cells to treat juvenile-onset diabetes.
Collapse
Affiliation(s)
- Warwick J Teague
- Nuffield Department of Surgery, University of Oxford, John Radcliffe Hospital, OX3 9DU Oxford, United Kingdom.
| | | | | | | |
Collapse
|
39
|
Abstract
This review discusses current understanding of the role that endogenous and exogenous progenitor cells may have in the treatment of the diseased heart. In the last several years, a major effort has been made in an attempt to identify immature cells capable of differentiating into cell lineages different from the organ of origin to be employed for the regeneration of the damaged heart. Embryonic stem cells (ESCs) and bone marrow-derived cells (BMCs) have been extensively studied and characterized, and dramatic advances have been made in the clinical application of BMCs in heart failure of ischemic and nonischemic origin. However, a controversy exists concerning the ability of BMCs to acquire cardiac cell lineages and reconstitute the myocardium lost after infarction. The recognition that the adult heart possesses a stem cell compartment that can regenerate myocytes and coronary vessels has raised the unique possibility to rebuild dead myocardium after infarction, to repopulate the hypertrophic decompensated heart with new better functioning myocytes and vascular structures, and, perhaps, to reverse ventricular dilation and wall thinning. Cardiac stem cells may become the most important cell for cardiac repair.
Collapse
Affiliation(s)
- Annarosa Leri
- Cardiovascular Research Institute, Department of Medicine, New York Medical College, Valhalla, NY10595, USA
| | | | | |
Collapse
|
40
|
Dickhut A, Schwerdtfeger R, Kuklick L, Ritter M, Thiede C, Neubauer A, Brendel C. Mesenchymal stem cells obtained after bone marrow transplantation or peripheral blood stem cell transplantation originate from host tissue. Ann Hematol 2005; 84:722-7. [PMID: 16132912 DOI: 10.1007/s00277-005-1067-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2004] [Accepted: 06/02/2005] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells (MSC) obtained from human bone marrow have been described as adult stem cells with the ability of extensive self-renewal and clonal expansion, as well as the capacity to differentiate into various tissue types and to modulate the immune system. Some data indicate that leukapheresis products may also contain non-hematopoietic stem cells, as they occur in whole bone marrow transplantation (BMT). However, there is still controversy whether MSC expand in the host after transplantation like blood progenitor cells do. Therefore, we were interested in finding out if graft MSC can be detected in leukapheresis products and in bone marrow after BMT and peripheral blood stem cell transplantation (PBSCT). Every sample from total bone marrow transplants exhibited growth of MSC after in vitro culture, but not one of nine leukapheresis products did. In addition, bone marrow aspirates of 9 patients receiving BMT and of 18 patients after PBSCT were examined for origin of MSC. Almost all MSC samples exhibited a complete host profile, whereas peripheral blood cells were of donor origin. We conclude that even if trace amounts of MSC are co-transplanted during PBSCT or BMT, they do not expand significantly in the host bone marrow.
Collapse
Affiliation(s)
- Andreas Dickhut
- Department of Hematology, Oncology and Immunology, Philipps-University of Marburg, 35033, Marburg, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
Zeng F, Chen M, Katsumata M, Huang W, Gong Z, Hu W, Qian H, Xiao Y, Ren Z, Huang S. Identification and characterization of engrafted human cells in human/goat xenogeneic transplantation chimerism. DNA Cell Biol 2005; 24:403-9. [PMID: 16008509 DOI: 10.1089/dna.2005.24.403] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have injected human CD34+lin- cells derived from cord blood (CB) into the goat fetuses via in utero at 45-55 days gestation under guidance of B-scan ultrasonograph. Sixty out of 68 fetuses injected survived to full term. The long-term survival of the human cells in transplant goat has been tested by various experimental methods, including FACS analysis, real-time PCR, RT-PCR, Southern-blot hybridization, FISH, as well as immunohistochemical assays. All the 60 transplant goats demonstrated engrafted human cells, including myeloid, B-lymphoid, and erythroid lineages. The yield of the human CD34+ cells varied, but was not linked with sex and age. High numbers of human cells could be detected for at least 16 months after birth. Immunohistochemical analyses revealed that the human cells were present not only in blood but also in other tissues, such as liver, of the transplant goats. In addition, a human-specific serum albumin and the hepatocyte nuclear factor (hHNF-3beta) mRNAs specific to human hepato-antigen could be readily detected in the livers of the transplant goats. Our results demonstrate that this in utero xenograft model should be useful for expansion of human HSC and possibly for the evaluating the effectiveness of prenatal treatment of human genetic diseases.
Collapse
Affiliation(s)
- Fanyi Zeng
- Shanghai Institute of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiao Tong University, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Muench MO. In utero transplantation: baby steps towards an effective therapy. Bone Marrow Transplant 2005; 35:537-47. [PMID: 15665844 DOI: 10.1038/sj.bmt.1704811] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In utero transplantation (IUT) offers the potential to treat a large number of diseases by transplantation of healthy cells into a fetus with a birth defect. Prenatal diagnosis is feasible for many diseases prior to the full development of the fetal immune system offering the opportunity to introduce foreign cells and antigens into the developing fetus. At least 45 cases of IUT have been performed for a variety of diseases. IUT has successfully treated severe combined immunodeficiency and there are indications that it may be effective in treating some nonhematopoietic diseases. However, many diseases remain resistant to fetal therapy owing to the low levels of chimerism that can be achieved. Promising efforts to improve the levels of engraftment are focusing on optimizing the graft and developing donor-specific tolerance in the fetal recipient. Mounting evidence suggests that donor T cells can aid in achieving clinically significant levels of chimerism. The use of fetal donor cells may also offer some benefit. Animal experiments suggest that even low-level chimerism can lead to tolerance, which can be exploited by booster transplants in the neonate. Continued research appears likely to succeed in developing IUT into an effective form of therapy for a variety of diseases.
Collapse
Affiliation(s)
- M O Muench
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143-0793, USA.
| |
Collapse
|
43
|
Prindull G. Hypothesis: Cell plasticity, linking embryonal stem cells to adult stem cell reservoirs and metastatic cancer cells? Exp Hematol 2005; 33:738-46. [PMID: 15963849 DOI: 10.1016/j.exphem.2005.03.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2005] [Revised: 02/24/2005] [Accepted: 03/02/2005] [Indexed: 11/26/2022]
Abstract
Embryonal stem (ES) cells are the earliest ontogenetically identifiable stem cells of the embryo proper for all subsequent mesenchymal stem cells and for highly specialized differentiated cells. This review characterizes, in a working hypothesis, the role of reversible EMT/MET (epithelialmesenchymal transition) as a manifestation of cell plasticity 1) in the development of ES cells to adult stem cells (hematopoietic stem cells) and 2) in metastasizing cancer cells. Animal studies support the concept that EMT/MET is a key manifestation of cell plasticity in the development of ES cells to adult stem cells, and in conversion of localized to metastasizing cancer cells. In fact, ES cells may persist to postnatal life, in cytologically verifiable form and/or within the frame of EMT/MET, as ultimate reservoir for adult stem cells. Furthermore, EMT could possibly serve as a conceptional link between physiologic and pathologic signaling pathways. Clonal confirmation in humans is necessary.
Collapse
Affiliation(s)
- Gregor Prindull
- Department of Pediatrics, University of Göttingen Medical School, Germany.
| |
Collapse
|
44
|
In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GMJS, Claas FHJ, Fibbe WE, Kanhai HHH. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 2005; 22:1338-45. [PMID: 15579651 DOI: 10.1634/stemcells.2004-0058] [Citation(s) in RCA: 812] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recently we reported that second-trimester amniotic fluid (AF) is an abundant source of fetal mesenchymal stem cells (MSCs). In this study, we analyze the origin of these MSCs and the presence of MSCs in human-term AF. In addition, different parts of the human placenta were studied for the presence of either fetal or maternal MSCs. We compared the phenotype and growth characteristics of MSCs derived from AF and placenta. Cells from human second-trimester (mean gestational age, 19(+2) [standard deviation, +/- 1(+3)] weeks, n = 10) and term third-trimester (mean gestational age, 38(+4) [standard deviation, +/- 1] weeks, n = 10) AF, amnion, decidua basalis, and decidua parietalis were cultured in M199 medium supplemented with 10% fetal calf serum and endothelial cell growth factor. Cultured cells were immunophenotypically characterized, the adipogenic and osteogenic differentiation capacity was tested, and the growth kinetics were analyzed. The origin of fetal and maternal cells was determined by molecular human leukocyte antigen typing. We successfully isolated MSCs from second-trimester AF, amnion, and decidua basalis as well as term amnion, decidua parietalis, and decidua basalis. In contrast, MSCs were cultured from only 2 out of 10 term AF samples. The phenotype of MSCs cultured from different fetal and maternal parts of the placenta was comparable. Maternal MSCs from second-trimester and term decidua basalis and parietalis showed a significantly higher expansion capacity than that of MSCs from adult bone marrow (p < .05). Our results indicate that both fetal and maternal MSCs can be isolated from the human placenta. Amnion is a novel source of fetal MSCs, likely contributing to the presence of MSCs in AF. Decidua basalis and decidua parietalis are sources for maternal MSCs. The expansion potency from both fetal and maternal placenta-derived MSCs was higher compared with adult bone marrow-derived MSCs.
Collapse
|
45
|
Santner-Nanan B, Peek MJ, McCullagh P, Nanan R. Therapeutic potential of stem cells in perinatal medicine. Aust N Z J Obstet Gynaecol 2005; 45:102-7. [PMID: 15760308 DOI: 10.1111/j.1479-828x.2005.00362.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Increasing evidence suggests that stem cells have tremendous potential to facilitate repair of damaged tissue and to exert protective influences that limit the extent of damage. Their inherent capacity to respond to signals generated by damaged tissue, migrate to these regions and either replace dead tissue or deliver protection by secretion of specific growth hormones and protective factors, suggests that they might have unrivalled therapeutic potential in perinatal medicine. A further potential of stem cells is their use in gene repair strategies for genetic disorders; an application which is exceedingly interesting from a perinatal perspective. Because of the relatively small size of infants and their capacity for future growth, stem cell therapy could be more successful in newborns than in older children or adults. In practical terms, the placenta, with its large reservoir of fetal blood, offers the ideal source of autologous stem cells. This affords the opportunity for stem cells to be collected and used, either directly ex vivo or after in vitro modulation, both for disorders in the neonatal period and for those arising later in life. The organs most affected from tissue damage in the neonatal period are the brain and the lung. So far, the most promising application of stem cells might be in the treatment of neurological injury. In this review we discuss recent research findings with adult stem cell therapy and their potential use in perinatal medicine. Furthermore, specific animal models suitable to explore the patho-physiological mechanisms of stem cell transplantation after neurological injury will be discussed. This review gives an overview of basic science findings and their possible role for clinical application with regards to the therapeutic potential of stem cells in perinatal medicine. Medline was searched for journal selection in peer-reviewed journals with high impact scores, which were relevant to this topic. All articles were in English and the search was not limited by publication year. However, the oldest publication was dated 1988 (reference 1).
Collapse
Affiliation(s)
- Brigitte Santner-Nanan
- Department of Women's and Children's Health, Western Clinical School, The University of Sydney, New South Wales, Australia
| | | | | | | |
Collapse
|
46
|
Abstract
Fetal stem cells can be isolated from fetal blood and bone marrow as well as from other fetal tissues, including liver and kidney. Fetal blood is a rich source of haemopoietic stem cells (HSC), which proliferate more rapidly than those in cord blood or adult bone marrow. First trimester fetal blood also contains a population of non-haemopoietic mesenchymal stem cells (MSC), which support haemopoiesis and can differentiate along multiple lineages. In terms of eventual downstream application, both fetal HSC and MSC have advantages over their adult counterparts, including better intrinsic homing and engraftment, greater multipotentiality and lower immunogenicity. Fetal stem cells are less ethically contentious than embryonic stem cells and their differentiation potential appears greater than adult stem cells. Fetal stem cells represent powerful tools for exploring many aspects of cell biology and hold considerable promise as therapeutic tools for cell transplantation and ex vivo gene therapy.
Collapse
Affiliation(s)
- Keelin O'Donoghue
- Experimental Fetal Medicine Group, Institute of Reproductive and Developmental Biology, Imperial College London, Queen Charlotte's and Chelsea Hospital, London W12 0NN, UK.
| | | |
Collapse
|
47
|
Fukuchi Y, Nakajima H, Sugiyama D, Hirose I, Kitamura T, Tsuji K. Human placenta-derived cells have mesenchymal stem/progenitor cell potential. Stem Cells 2005; 22:649-58. [PMID: 15342929 DOI: 10.1634/stemcells.22-5-649] [Citation(s) in RCA: 433] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mesenchymal stem/progenitor cells (MSCs) are widely distributed in a variety of tissues in the adult human body (e.g., bone marrow [BM], kidney, lung, and liver). These cells are also present in the fetal environment (e.g., blood, liver, BM, and kidney). However, MSCs are a rare population in these tissues. Here we tried to identify cells with MSC-like potency in human placenta. We isolated adherent cells from trypsin-digested term placentas and established two clones by limiting dilution. We examined these cells for morphology, surface markers, gene expression patterns, and differentiation potential and found that they expressed several stem cell markers, hematopoietic/ endothelial cell-related genes, and organ-specific genes, as determined by reverse transcription-polymerase chain reaction and fluorescence-activated cell sorter analysis. They also showed osteogenic and adipogenic differentiation potentials under appropriate conditions. We suggest that placenta-derived cells have multilineage differentiation potential similar to MSCs in terms of morphology, cell-surface antigen expression, and gene expression patterns. The placenta may prove to be a useful source of MSCs.
Collapse
Affiliation(s)
- Yumi Fukuchi
- Division of Cellular Therapy, The Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639, Japan.
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Kidney transplantation has been one of the major medical advances of the past 30 years; however, it is becoming increasingly apparent that the supply of organs is limited and will not improve with current medical practice. This review summarizes recent data whereby precursors of the adult kidney found in embryos and fetal tissue have been grafted into murine hosts to examine their feasibility as an alternative source for renal transplantation. When obtained at specific time points during human gestation, kidney precursors meet with specific demands; they grow tremendously, differentiate exclusively along the nephric lineage with no evidence of malignant transformation, become vascularised, to a larger extent, by host vessels, and produce urine in host animals. In addition, they exhibit decreased immunogenicity compared to adult counterparts. Organogenesis is best achieved when utilizing early undifferentiated progenitors rather than later-gestation kidneys. Nevertheless, in order for these transplants to be applicable for human transplantation, both a functional urinary anastomosis and derivation of blood supply sufficient to correct biochemical abnormalities remain to be established.
Collapse
Affiliation(s)
- Benjamin Dekel
- Developmental Biology Laboratory, Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.
| | | |
Collapse
|
49
|
Dekel B, Reisner Y. Embryonic committed stem cells as a solution to kidney donor shortage. Expert Opin Biol Ther 2004; 4:443-54. [PMID: 15102595 DOI: 10.1517/14712598.4.4.443] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The number of human kidney transplants has increased rapidly in recent years, but the need greatly exceeds organ availability. Induction of appropriate kidney differentiation and growth from stem or progenitor cell populations represents an attractive option to combat chronic kidney donor shortage. In an analogy to haematopoietic stem cells, which are much more efficient in giving rise to blood than to other cell types, if any at all, renal stem cells could afford an unlimited source for regenerating nephrons. While a single nephrogenic stem cell has not been characterised, indirect evidence suggests that a renal stem cell population is contained within the metanephric mesenchyme, which along with a branch of the Wolffian duct represents the direct precursor of the mature kidney. Human tissue fragments derived from these developing precursors can regenerate renal structures when grafted into mice. Moreover, recent data pinpoints a window of time in human and pig kidney development that may be optimal for transplantation into mature recipients. 'Window' transplants are defined by their remarkable ability to grow, differentiate and undergo vascularisation, achieving successful organogenesis of urine-producing miniature kidneys with no evidence of transdifferentiation into non-renal cell types, lack of tumourigenicity and reduced immunogenicity compared with adult counterparts. In contrast, 'non-window' transplants (earlier or later in gestation) can form teratomas or are more prone to immune rejection and are less suitable for organogenesis. Hopefully, the use of stage-specific early human and porcine kidney precursors to cultivate mature kidney cells in vivo, possibly in conjunction with other modalities of stem cell technology and tissue engineering, will prove valuable to sustain life in patients with failing kidneys.
Collapse
Affiliation(s)
- Benjamin Dekel
- Weizmann Institute of Science, Department of Immunology, Rehovot, Israel
| | | |
Collapse
|
50
|
Almeida-Porada G, Porada CD, Chamberlain J, Torabi A, Zanjani ED. Formation of human hepatocytes by human hematopoietic stem cells in sheep. Blood 2004; 104:2582-90. [PMID: 15231580 DOI: 10.1182/blood-2004-01-0259] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We took advantage of the proliferative and permissive environment of the developing preimmune fetus to develop a noninjury large animal model in sheep, in which the transplantation of defined populations of human hematopoietic stem cells resulted in the establishment of human hematopoiesis and led to the formation of significant numbers of long-lasting, functional human liver cells, with some animals exhibiting levels as high as 20% of donor (human) hepatocytes 11 months after transplantation. A direct correlation was found between hepatocyte activity and phenotype of transplanted cells, cell dose administered, source of cells used on a cell-per-cell basis (bone marrow, cord blood, mobilized peripheral blood), and time after transplantation. Human hepatocytes generated in this model retained functional properties of normal hepatocytes, constituted hepatic functional units with the presence of human endothelial and biliary duct cells, and secreted human albumin that was detected in circulation. Transplanting populations of hematopoietic stem cells can efficiently generate significant numbers of functional hepatic cells in this noninjury large animal model and thus could be a means of ameliorating or curing genetic diseases in which a deficiency of liver cells or their products threatens the life of the fetus or newborn.
Collapse
Affiliation(s)
- Graça Almeida-Porada
- Department of Animal Biotechnology, University of Nevada Reno, Mail Stop 202, Reno, NV 89557-0104, USA.
| | | | | | | | | |
Collapse
|