1
|
Kihara Y, Tanaka Y, Ikeda M, Homma J, Takagi R, Ishigaki K, Yamanouchi K, Honda H, Nagata S, Yamato M. In utero transplantation of myoblasts and adipose-derived mesenchymal stem cells to murine models of Duchenne muscular dystrophy does not lead to engraftment and frequently results in fetal death. Regen Ther 2022; 21:486-493. [PMID: 36313392 PMCID: PMC9596598 DOI: 10.1016/j.reth.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/19/2022] [Accepted: 10/09/2022] [Indexed: 11/07/2022] Open
Abstract
Introduction Duchenne muscular dystrophy (DMD) is a progressive disease that leads to damage of muscle and myocardium due to genetic abnormalities in the dystrophin gene. In utero cell transplantation that might facilitate allogenic transplantation is worth considering to treat this disease. Methods We performed allogeneic in utero transplantation of GFP-positive myoblasts and adipose-derived mesenchymal stem cells into murine DMD model animals. The transplantation route in this study was fetal intraperitoneal transplantation and transplacental transplantation. Transplanted animals were examined at 4-weeks old by immunofluorescence staining and RT-qPCR. Results No GFP-positive cells were found by immunofluorescence staining of skeletal muscle and no GFP mRNA was detected by RT-qPCR in any animal, transplantation method and cell type. Compared with previous reports, myoblast transplantation exhibited an equivalent mortality rate, but adipose-derived stem cell (ASC) transplantation produced a higher mortality rate. Conclusions In utero transplantation of myoblasts or ASCs to murine models of DMD does not lead to engraftment and, in ASC transplantation primarily, frequently results in fetal death.
Collapse
Affiliation(s)
- Yuki Kihara
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan,Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Yukie Tanaka
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Masanari Ikeda
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Jun Homma
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Ryo Takagi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Keiko Ishigaki
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Hiroaki Honda
- Human Disease Models, Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Satoru Nagata
- Department of Pediatrics, Tokyo Women’s Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan,Corresponding author. Fax: +81 3-3359-6046.
| |
Collapse
|
2
|
Shi C, Pan L, Hu Z. Experimental and clinical progress of in utero hematopoietic cell transplantation therapy for congenital disorders. Front Pharmacol 2022; 13:851375. [PMID: 36120324 PMCID: PMC9478511 DOI: 10.3389/fphar.2022.851375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
In utero hematopoietic cell transplantation (IUHCT) is considered a potentially efficient therapeutic approach with relatively few side effects, compared to adult hematopoietic cell transplantation, for various hematological genetic disorders. The principle of IUHCT has been extensively studied in rodent models and in some large animals with close evolutionary similarities to human beings. However, IUHCT has only been used to rebuild human T cell immunity in certain patients with inherent immunodeficiencies. This review will first summarize the animal models utilized for IUHCT investigations and describe the associated outcomes. Recent advances and potential barriers for successful IUHCT are discussed, followed by possible strategies to overcome these barriers experimentally. Lastly, we will outline the progress made towards utilizing IUHCT to treat inherent disorders for patients, list out associated limitations and propose feasible means to promote the efficacy of IUHCT clinically.
Collapse
Affiliation(s)
- Chunyu Shi
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lu Pan
- Department of Pediatric Immunology, Allergy and Rheumatology, The First Hospital of Jilin University, Changchun, China
| | - Zheng Hu
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Zheng Hu,
| |
Collapse
|
3
|
Boss AL, Damani T, Wickman TJ, Chamley LW, James JL, Brooks AES. Full spectrum flow cytometry reveals mesenchymal heterogeneity in first trimester placentae and phenotypic convergence in culture, providing insight into the origins of placental mesenchymal stromal cells. eLife 2022; 11:76622. [PMID: 35920626 PMCID: PMC9371602 DOI: 10.7554/elife.76622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 08/01/2022] [Indexed: 12/05/2022] Open
Abstract
Single-cell technologies (RNA-sequencing, flow cytometry) are critical tools to reveal how cell heterogeneity impacts developmental pathways. The placenta is a fetal exchange organ, containing a heterogeneous mix of mesenchymal cells (fibroblasts, myofibroblasts, perivascular, and progenitor cells). Placental mesenchymal stromal cells (pMSC) are also routinely isolated, for therapeutic and research purposes. However, our understanding of the diverse phenotypes of placental mesenchymal lineages, and their relationships remain unclear. We designed a 23-colour flow cytometry panel to assess mesenchymal heterogeneity in first-trimester human placentae. Four distinct mesenchymal subsets were identified; CD73+CD90+ mesenchymal cells, CD146+CD271+ perivascular cells, podoplanin+CD36+ stromal cells, and CD26+CD90+ myofibroblasts. CD73+CD90+ and podoplanin + CD36+ cells expressed markers consistent with cultured pMSCs, and were explored further. Despite their distinct ex-vivo phenotype, in culture CD73+CD90+ cells and podoplanin+CD36+ cells underwent phenotypic convergence, losing CD271 or CD36 expression respectively, and homogenously exhibiting a basic MSC phenotype (CD73+CD90+CD31-CD144-CD45-). However, some markers (CD26, CD146) were not impacted, or differentially impacted by culture in different populations. Comparisons of cultured phenotypes to pMSCs further suggested cultured pMSCs originate from podoplanin+CD36+ cells. This highlights the importance of detailed cell phenotyping to optimise therapeutic capacity, and ensure use of relevant cells in functional assays.
Collapse
Affiliation(s)
- Anna Leabourn Boss
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Tanvi Damani
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Tayla J Wickman
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Larry W Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Jo L James
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Anna E S Brooks
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
4
|
Stokes SC, Theodorou CM, Jackson JE, Pivetti C, Kumar P, Yamashiro KJ, Paxton ZJ, Reynaga L, Hyllen A, Wang A, Farmer DL. Long-term safety evaluation of placental mesenchymal stromal cells for in utero repair of myelomeningocele in a novel ovine model. J Pediatr Surg 2022; 57:18-25. [PMID: 34657738 PMCID: PMC9415987 DOI: 10.1016/j.jpedsurg.2021.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE Augmentation of in utero myelomeningocele repair with human placental mesenchymal stromal cells seeded onto extracellular matrix (PMSC-ECM) improves motor outcomes in an ovine myelomeningocele model. This study evaluated the safety of PMSC-ECM application directly onto the fetal spinal cord in preparation for a clinical trial. METHODS Laminectomy of L5-L6 with PMSC-ECM placement directly onto the spinal cord was performed in five fetal lambs at gestational age (GA) 100-106 days. Lambs and ewes were monitored for three months following delivery. Lambs underwent magnetic resonance imaging (MRI) of the brain and spine at birth and at three months. All organs from lambs and uteri from ewes underwent histologic evaluation. Lamb spinal cords and brains and ewe placentas were evaluated for persistence of PMSCs by polymerase chain reaction for presence of human DNA. RESULTS MRIs demonstrated no evidence of abnormal tissue growth or spinal cord tethering. Histological analysis demonstrated no evidence of abnormal tissue growth or treatment related adverse effects. No human DNA was identified in evaluated tissues. CONCLUSION There was no evidence of abnormal tissue growth or PMSC persistence at three months following in utero application of PMSC-ECM to the spinal cord. This supports proceeding with clinical trials of PMSC-ECM for in utero myelomeningocele repair. LEVEL OF EVIDENCE N/A TYPE OF STUDY: Basic science.
Collapse
Affiliation(s)
- Sarah C Stokes
- Division of Pediatric General, Thoracic and Fetal Surgery, Department of Surgery, University of California Davis Medical Center, 2335 Stockton Blvd, Room 5107, Sacramento, CA 95817, USA.
| | - Christina M Theodorou
- Division of Pediatric General, Thoracic, and Fetal Surgery, Department of Surgery, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Jordan E Jackson
- Division of Pediatric General, Thoracic, and Fetal Surgery, Department of Surgery, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Christopher Pivetti
- Surgical Bioengineering Laboratory, University of California Davis, Sacramento, CA 95817, USA
| | - Priyadarsini Kumar
- Surgical Bioengineering Laboratory, University of California Davis, Sacramento, CA 95817, USA
| | - Kaeli J Yamashiro
- Division of Pediatric General, Thoracic, and Fetal Surgery, Department of Surgery, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Zachary J Paxton
- Surgical Bioengineering Laboratory, University of California Davis, Sacramento, CA 95817, USA
| | - Lizette Reynaga
- Surgical Bioengineering Laboratory, University of California Davis, Sacramento, CA 95817, USA
| | - Alicia Hyllen
- Surgical Bioengineering Laboratory, University of California Davis, Sacramento, CA 95817, USA
| | - Aijun Wang
- Surgical Bioengineering Laboratory, University of California Davis, Sacramento, CA 95817, USA.,Shriners Hospital for Children Northern California, 3425 Stockton Blvd, Sacramento, CA 95817, USA
| | - Diana L Farmer
- Division of Pediatric General, Thoracic, and Fetal Surgery, Department of Surgery, University of California Davis Medical Center, Sacramento, CA 95817, USA.,Surgical Bioengineering Laboratory, University of California Davis, Sacramento, CA 95817, USA.,Shriners Hospital for Children Northern California, 3425 Stockton Blvd, Sacramento, CA 95817, USA
| |
Collapse
|
5
|
Lu Y, Zhou Y, Ju R, Chen J. Human-animal chimeras for autologous organ transplantation: technological advances and future perspectives. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:576. [PMID: 31807557 DOI: 10.21037/atm.2019.10.13] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Organ transplantation is the most promising curation for end-stage organ disease. However, the donor organ shortage has become a global problem that has limited the development of organ transplantation. Human-animal chimeras provide the ability to produce human organs in other species using autologous stem cells [e.g., induced pluripotent stem cells (iPSCs) or adult stem cells], which would be patient-specific and immune-matched for transplantation. Due to the potential application prospect of interspecies chimeras in basic and translational research, this technology has attracted much interest. This review focuses primarily on technological advances, including options of donor stem cell types and gene editing in donor cells and host animals, in addition to perspectives on human-animal chimeras in clinical and basic research.
Collapse
Affiliation(s)
- Yingfei Lu
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Yu Zhou
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China.,Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Rong Ju
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China.,Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| |
Collapse
|
6
|
Zhang Y, Xu J, Liu S, Lim M, Zhao S, Cui K, Zhang K, Wang L, Ji Q, Han Z, Kong D, Li Z, Liu N. Embryonic stem cell-derived extracellular vesicles enhance the therapeutic effect of mesenchymal stem cells. Am J Cancer Res 2019; 9:6976-6990. [PMID: 31660081 PMCID: PMC6815953 DOI: 10.7150/thno.35305] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/29/2019] [Indexed: 01/04/2023] Open
Abstract
Background: Embryonic stem cells (ES) have a great potential for cell-based therapies in a regenerative medicine. However, the ethical and safety issues limit its clinical application. ES-derived extracellular vesicles (ES-EVs) have been reported suppress cellular senescence. Mesenchymal stem cells (MSCs) are widely used for clinical cell therapy. In this study, we investigated the beneficial effects of ES-EVs on aging MSCs to further enhancing their therapeutic effects. Methods: In vitro, we explored the rejuvenating effects of ES-EVs on senescent MSCs by senescence-associated β-gal (SA-β-gal) staining, immunostaining, and DNA damage foci analysis. The therapeutic effect of senescent MSC pre-treated with ES-EVs was also evaluated by using mouse cutaneous wound model. Results: We found that ES-EVs significantly rejuvenated the senescent MSCs in vitro and improve the therapeutic effects of MSCs in a mouse cutaneous wound model. In addition, we also identified that the IGF1/PI3K/AKT pathway mediated the antisenescence effects of ES-EVs on MSCs. Conclusions: Our results suggested that ES cells derived-extracellular vesicles possess the antisenescence properties, which significantly rejuvenate the senescent MSCs and enhance the therapeutic effects of MSCs. This strategy might emerge as a novel therapeutic strategy for MSCs clinical application.
Collapse
|
7
|
Muñoz-Fernández R, De La Mata C, Requena F, Martín F, Fernandez-Rubio P, Llorca T, Ruiz-Magaña MJ, Ruiz-Ruiz C, Olivares EG. Human predecidual stromal cells are mesenchymal stromal/stem cells and have a therapeutic effect in an immune-based mouse model of recurrent spontaneous abortion. Stem Cell Res Ther 2019; 10:177. [PMID: 31200769 PMCID: PMC6567662 DOI: 10.1186/s13287-019-1284-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022] Open
Abstract
Background Human decidual stromal cells (DSCs) are involved in the maintenance and development of pregnancy, in which they play a key role in the induction of immunological maternal–fetal tolerance. Precursors of DSCs (preDSCs) are located around the vessels, and based on their antigen phenotype, previous studies suggested a relationship between preDSCs and mesenchymal stromal/stem cells (MSCs). This work aimed to further elucidate the MSC characteristics of preDSCs. Methods We established 15 human preDSC lines and 3 preDSC clones. Physiological differentiation (decidualization) of these cell lines and clones was carried out by in vitro culture with progesterone (P4) and cAMP. Decidualization was confirmed by the change in cellular morphology and prolactin (PRL) secretion, which was determined by enzyme immunoassay of the culture supernatants. We also studied MSC characteristics: (1) In mesenchymal differentiation, under appropriate culture conditions, these preDSC lines and clones differentiated into adipocytes, osteoblasts, and chondrocytes, and differentiation was confirmed by cytochemical assays and RT-PCR. (2) The expression of stem cell markers was determined by RT-PCR. (3) Cloning efficiency was evaluated by limited dilution. (4) Immunoregulatory activity in vivo was estimated in DBA/2-mated CBA/J female mice, a murine model of immune-based recurrent abortion. (5) Survival of preDSC in immunocompetent mice was analyzed by RT-PCR and flow cytometry. Results Under the effect of P4 and cAMP, the preDSC lines and clones decidualized in vitro: the cells became rounder and secreted PRL, a marker of physiological decidualization. PreDSC lines and clones also exhibited MSC characteristics. They differentiated into adipocytes, osteoblasts, and chondrocytes, and preDSC lines expressed stem cell markers OCT-4, NANOG, and ABCG2; exhibited a cloning efficiency of 4 to 15%; significantly reduced the embryo resorption rate (P < 0.001) in the mouse model of abortion; and survived for prolonged periods in immunocompetent mice. The fact that 3 preDSC clones underwent both decidualization and mesenchymal differentiation shows that the same type of cell exhibited both DSC and MSC characteristics. Conclusions Together, our results confirm that preDSCs are decidual MSCs and suggest that these cells are involved in the mechanisms of maternal–fetal immune tolerance. Electronic supplementary material The online version of this article (10.1186/s13287-019-1284-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raquel Muñoz-Fernández
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Claudia De La Mata
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Francisco Requena
- Departamento de Estadística e Investigación Operativa, Universidad de Granada, Granada, Spain
| | - Francisco Martín
- Human DNA Variability Department, GENYO - Centre for Genomic and Oncological Research (Pfizer/University of Granada/Andalusian Regional Government), PTS Granada, Granada, Spain
| | - Pablo Fernandez-Rubio
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Tatiana Llorca
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Maria José Ruiz-Magaña
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Carmen Ruiz-Ruiz
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Avenida de la Investigación, 11, 18016, Granada, Spain
| | - Enrique G Olivares
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain. .,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Avenida de la Investigación, 11, 18016, Granada, Spain. .,Unidad de Gestión Clínica Laboratorios, Hospital Universitario San Cecilio, Granada, Spain.
| |
Collapse
|
8
|
Boss AL, Chamley LW, James JL. Placental formation in early pregnancy: how is the centre of the placenta made? Hum Reprod Update 2019; 24:750-760. [PMID: 30257012 DOI: 10.1093/humupd/dmy030] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/09/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Correct development of the placenta is critical to establishing pregnancy and inadequate placentation leads to implantation failure and miscarriage, as well as later gestation pregnancy disorders. Much attention has been focused on the placental trophoblasts and it is clear that the trophoblast lineages arise from the trophectoderm of the blastocyst. In contrast, the cells of the placental mesenchyme are thought to arise from the inner cell mass, but the details of this process are limited. Due to ethical constraints and the inaccessibility of very early implantation tissues, our knowledge of early placentation has been largely based on historical histological sections. More recently, stem cell technologies have begun to shed important new light on the origins of the placental mesenchymal lineages. OBJECTIVE AND RATIONALE This review aims to amalgamate the older and more modern literature regarding the origins of the non-trophoblast lineages of the human placenta. We highlight ways in which rapidly developing stem cell technologies may shed new light on these crucial peri-implantation events. SEARCH METHODS Relevant articles were identified using the PubMed database and Google Scholar search engines. A pearl growing method was used to expand the scope of papers relevant to the cell differentiation events of non-trophoblast placental lineages. OUTCOMES At the start of pregnancy, cells of the extraembyronic mesoderm migrate to underlie the primitive trophoblast layers forming the first placental villi. The mesenchymal cells in the villus core most likely originate from the hypoblast of the embryo, but whether cells from the epiblast also contribute is yet to be determined. This is important because, following the formation of the villus core, vasculogenesis and haematopoiesis take place in the nascent placenta before it is connected to the embryonic circulation, making it likely that haematopoietic foci, placental macrophages, endothelial cells and vascular smooth muscle cells all arise in the placenta de novo. Evidence from the stem cell field indicates that these cells could directly differentiate from the extraembryonic mesoderm. However, the lineage hierarchy involved in cell fate decisions has not been well-established. Mesodermal progenitors capable of differentiating into both vascular and haematopoietic lineages can be derived from human embryonic stem cells, but the identification of such stem cells in the placenta is lacking. Future work profiling rare progenitor populations in early placentae will aid our understanding of early placentation. WIDER IMPLICATIONS Understanding the origins of the cell lineages of the normal placenta will help us understand why so many pregnancies fail and address mechanisms that may salvage some of these losses.
Collapse
Affiliation(s)
- Anna L Boss
- Department of Obstetrics and Gynecology, University of Auckland, 85 Park Rd, Grafton, Auckland, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynecology, University of Auckland, 85 Park Rd, Grafton, Auckland, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynecology, University of Auckland, 85 Park Rd, Grafton, Auckland, New Zealand
| |
Collapse
|
9
|
Kumar P, Gao K, Wang C, Pivetti C, Lankford L, Farmer D, Wang A. In Utero Transplantation of Placenta-Derived Mesenchymal Stromal Cells for Potential Fetal Treatment of Hemophilia A. Cell Transplant 2019; 27:130-139. [PMID: 29562772 PMCID: PMC6434487 DOI: 10.1177/0963689717728937] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hemophilia A (HA) is an X-linked recessive disorder caused by mutations in the factor VIII (FVIII) gene leading to deficient blood coagulation. The current standard of care is frequent infusions of plasma-derived FVIII or recombinant B-domain-deleted FVIII (BDD-FVIII). While this treatment is effective, many patients eventually develop FVIII inhibitors that limit the effectiveness of the infused FVIII. As a monogenic disorder, HA is an ideal target for gene or cell-based therapy. Several studies have investigated allogeneic stem cell therapy targeting in utero or postnatal treatment of HA but have not been successful in completely correcting HA. Autologous in utero transplantation of mesenchymal stem cells is promising for treatment of HA due to the naive immune status of the fetal environment as well as its potential to prevent transplant rejection and long-term FVIII inhibitor formation. HA can be diagnosed by chorionic villus sampling performed during the first trimester (10 to 13 wk) of gestation. In this study, we used an established protocol and isolated placenta-derived mesenchymal stromal cells (PMSCs) from first trimester chorionic villus tissue and transduced them with lentiviral vector encoding the BDD-FVIII gene. We show that gene-modified PMSCs maintain their immunophenotype and multipotency, express, and secrete high levels of active FVIII. PMSCs were then transplanted at embryonic day 14.5 (E14.5) into wild-type fetuses from time-mated pregnant mice. Four days after birth, pups were checked for engraftment, and varying levels of expression of human green fluorescent protein were found in the organs tested. This study shows feasibility of the approach to obtain PMSCs from first trimester chorionic villus tissue, genetically modify them with the FVIII gene, and transplant them in utero for cell-mediated gene therapy of HA. Future studies will involve evaluation of long-term engraftment, phenotypic correction in HA mice, and prevention of FVIII inhibitor development by this approach.
Collapse
Affiliation(s)
- Priyadarsini Kumar
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Kewa Gao
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA.,2 Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Chuwang Wang
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA.,2 Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Christopher Pivetti
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Lee Lankford
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Diana Farmer
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| | - Aijun Wang
- 1 Department of Surgery, Surgical Bioengineering Laboratory, UC Davis School of Medicine, Research II, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
10
|
Teng CF, Jeng LB, Shyu WC. Role of Insulin-like Growth Factor 1 Receptor Signaling in Stem Cell Stemness and Therapeutic Efficacy. Cell Transplant 2018; 27:1313-1319. [PMID: 29882416 PMCID: PMC6168993 DOI: 10.1177/0963689718779777] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Evidence has emerged that stem cells represent a promising therapeutic tool for tissue engineering and regenerative medicine. Thus, identifying functional markers for selecting stem cells capable of superior self-renewal and pluripotency (or multipotency) and maintaining stem cell identity under appropriate culture conditions are critical for guiding the use of stem cells toward clinical applications. Many investigations have implicated the insulin-like growth factor 1 receptor (IGF1R) signaling in maintenance of stem cell characteristics and enhancement of stem cell therapy efficacy. IGF1R-expressing stem cells display robust pluripotent or multipotent properties. In this review, we summarize the essential roles of IGF1R signaling in self-renewal, pluripotency (or multipotency), and therapeutic efficacy of stem cells, including human embryonic stem cells, neural stem cells, cardiac stem cells, bone marrow mesenchymal stem cells, placental mesenchymal stem cells, and dental pulp mesenchymal stem cells. Modifying IGF1R signaling may thus provide potential strategies for maintaining stem cell properties and improving stem-cell-based therapeutic applications.
Collapse
Affiliation(s)
- Chiao-Fang Teng
- 1 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,2 Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Long-Bin Jeng
- 2 Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Woei-Cherng Shyu
- 1 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,3 Translational Medicine Research Center and Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,4 Department of Occupational Therapy, Asia University, Taichung, Taiwan
| |
Collapse
|
11
|
Potential clinical applications of placental stem cells for use in fetal therapy of birth defects. Placenta 2017; 59:107-112. [PMID: 28651900 DOI: 10.1016/j.placenta.2017.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/22/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
Placental stem cells are of growing interest for a variety of clinical applications due to their multipotency and ready availability from otherwise frequently discarded biomaterial. Stem cells derived from the placenta have been investigated in a number of disease processes, including wound healing, ischemic heart disease, autoimmune disorders, and chronic lung or liver injury. Fetal intervention for structural congenital defects, such as spina bifida, has rapidly progressed as a field due to advances in maternal-fetal medicine and improving surgical techniques. In utero treatment of structural, as well as non-structural, congenital disorders with cell-based therapies is of particular interest given the immunologic immaturity and immunotolerant environment of the developing fetus. A comprehensive literature review was performed to assess the potential utilization of placenta-derived stem cells for in utero treatment of congenital disorders. Most studies are still in the preclinical phase, utilizing animal models of common congenital disorders. Future research endeavors may include autologous transplantation, gene transfers, induced pluripotent stem cells, or cell-free therapies derived from the stem cell secretome. Though much work still needs to be done, placental stem cells are a promising therapeutic agent for fetal intervention for congenital disease.
Collapse
|
12
|
In Utero Stem Cell Transplantation: Potential Therapeutic Application for Muscle Diseases. Stem Cells Int 2017; 2017:3027520. [PMID: 28596791 PMCID: PMC5450178 DOI: 10.1155/2017/3027520] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/26/2017] [Indexed: 01/07/2023] Open
Abstract
Muscular dystrophies, myopathies, and traumatic muscle injury and loss encompass a large group of conditions that currently have no cure. Myoblast transplantations have been investigated as potential cures for these conditions for decades. However, current techniques lack the ability to generate cell numbers required to produce any therapeutic benefit. In utero stem cell transplantation into embryos has been studied for many years mainly in the context of hematopoietic cells and has shown to have experimental advantages and therapeutic applications. Moreover, patient-derived cells can be used for experimental transplantation into nonhuman animal embryos via in utero injection as the immune response is absent at such early stages of development. We therefore propose in utero transplantation as a potential method to generate patient-derived humanized skeletal muscle as well as muscle stem cells in animals for therapeutic purposes as well as patient-specific drug screening.
Collapse
|
13
|
Sardesai VS, Shafiee A, Fisk NM, Pelekanos RA. Avoidance of Maternal Cell Contamination and Overgrowth in Isolating Fetal Chorionic Villi Mesenchymal Stem Cells from Human Term Placenta. Stem Cells Transl Med 2017; 6:1070-1084. [PMID: 28205414 PMCID: PMC5442838 DOI: 10.1002/sctm.15-0327] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 07/05/2016] [Accepted: 07/28/2016] [Indexed: 12/16/2022] Open
Abstract
Human placenta is rich in mesenchymal stem/stromal cells (MSC), with their origin widely presumed fetal. Cultured placental MSCs are confounded by a high frequency of maternal cell contamination. Our recent systematic review concluded that only a small minority of placental MSC publications report fetal/maternal origin, and failed to discern a specific methodology for isolation of fetal MSC from term villi. We determined isolation conditions to yield fetal and separately maternal MSC during ex vivo expansion from human term placenta. MSCs were isolated via a range of methods in combination; selection from various chorionic regions, different commercial media, mononuclear cell digest and/or explant culture. Fetal and maternal cell identities were quantitated in gender‐discordant pregnancies by XY chromosome fluorescence in situ hybridization. We first demonstrated reproducible maternal cell contamination in MSC cultures from all chorionic anatomical locations tested. Cultures in standard media rapidly became composed entirely of maternal cells despite isolation from fetal villi. To isolate pure fetal cells, we validated a novel isolation procedure comprising focal dissection from the cotyledonary core, collagenase/dispase digestion and explant culture in endothelial growth media that selected, and provided a proliferative environment, for fetal MSC. Comparison of MSC populations within the same placenta confirmed fetal to be smaller, more osteogenic and proliferative than maternal MSC. We conclude that in standard media, fetal chorionic villi‐derived MSC (CV‐MSC) do not grow readily, whereas maternal MSC proliferate to result in maternal overgrowth during culture. Instead, fetal CV‐MSCs require isolation under specific conditions, which has implications for clinical trials using placental MSC. Stem Cells Translational Medicine2017;6:1070–1084
Collapse
Affiliation(s)
- Varda S Sardesai
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia
| | - Abbas Shafiee
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Nicholas M Fisk
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia.,Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - Rebecca A Pelekanos
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia
| |
Collapse
|
14
|
Chen CY, Tsai CH, Chen CY, Wu YH, Chen CP. Human placental multipotent mesenchymal stromal cells modulate placenta angiogenesis through Slit2-Robo signaling. Cell Adh Migr 2016; 10:66-76. [PMID: 26745454 PMCID: PMC4853036 DOI: 10.1080/19336918.2015.1108510] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/07/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022] Open
Abstract
The objective of this study was to investigate whether human placental multipotent mesenchymal stromal cell (hPMSC)-derived Slit2 and endothelial cell Roundabout (Robo) receptors are involved in placental angiogenesis. The hPMSC-conditioned medium and human umbilical vein endothelial cells were studied for Slit2 and Robo receptor expression by immunoassay and RT-PCR. The effect of the conditioned medium of hPMSCs with or without Slit2 depletion on endothelial cells was investigated by in vitro angiogenesis using growth factor-reduced Matrigel. hPMSCs express Slit2 and both Robo1 and Robo4 are present in human umbilical vein endothelial cells. Human umbilical vein endothelial cells do not express Robo2 and Robo3. The hPMSC-conditioned medium and Slit2 recombinant protein significantly inhibit the endothelial cell migration, but not by the hPMSC-conditioned medium with Slit2 depletion. The hPMSC-conditioned medium and Slit2 significantly enhance endothelial tube formation with increased cumulated tube length, polygonal network number and vessel branching point number compared to endothelial cells alone. The tube formation is inhibited by the depletion of Slit2 from the conditioned medium, or following the expression of Robo1, Robo4, and both receptor knockdown using small interfering RNA. Furthermore, co-immunoprecipitation reveals Slit2 binds to Robo1 and Robo4. Robo1 interacts and forms a heterodimeric complex with Robo4. These results suggest the implication of both Robo receptors with Slit2 signaling, which is involved in endothelial cell angiogenesis. Slit2 in the conditioned medium of hPMSCs has functional effect on endothelial cells and may play a role in placental angiogenesis.
Collapse
Affiliation(s)
- Cheng-Yi Chen
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chin-Han Tsai
- Division of High Risk Pregnancy, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chia-Yu Chen
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Hsin Wu
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chie-Pein Chen
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Division of High Risk Pregnancy, MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
15
|
Zhang W, Yang H, Zhang Y, Lu Y, Zhou T, Li M, Wen Y, Lin X, Xiang R, Chen X. A biotherapy based on PSCs-in-3D spheroid-ameliorated biologics depletes in vivo cancer-sustaining stem cells. Oncotarget 2015; 6:40762-74. [PMID: 26512920 PMCID: PMC4747367 DOI: 10.18632/oncotarget.5691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 09/14/2015] [Indexed: 02/05/2023] Open
Abstract
CSCs are able to survive routine anticancer procedures and peripheral-immune attack. Here we develop and detail a framework of CSC elimination governed by 3D-biologics. Pluripotent cells-engineered 3D-biologics (PMSB) and control non-3D-biologics were prepared from placenta-based somatic stem cells (PSCs) and inoculated respectively into senile hosts bearing progressive mammary, lung, colon carcinomas and melanoma. We demonstrate that PMSB evokes in vivo central-immune microenvironment with subsequent re-expression of thymosin-α1 ~ β4 in thymic cortex-medulla borderline for rapid MHC-unrestricted renewal of γδT-dominated immunocompetence. The post-renewal γδT-subsets could accurately bind and drive CSCs into apoptosis. Finally, with central/peripheral integral microenvironment renewal and TERT/Wnt/β-catenin pathway blockade, the CSC-subsets are fully depleted, leading to substantial cure of diverse tumors by PMSB inoculation (P < 0.01), yet not by non-3D-biologics. Thus, our study may contribute to open up a new avenue for tumor remission via pluripotent cells-engineered 3D-biologics addressing quick renewal of central-thymus and peripheral immune-microenvironment.
Collapse
Affiliation(s)
- Wenhui Zhang
- National Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Huanhuan Yang
- National Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yanna Zhang
- National Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yanan Lu
- National Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Tianlin Zhou
- National Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Meng Li
- National Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yanjun Wen
- Department of Gynecology & Obstetrics, West China Hospital/Second Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xiaojuan Lin
- Department of Gynecology & Obstetrics, West China Hospital/Second Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Rong Xiang
- Department of Immunology, Nankai University School of Medicine, Tianjin, People's Republic of China
| | - Xiancheng Chen
- National Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
16
|
Chen CY, Liu SH, Chen CY, Chen PC, Chen CP. Human placenta-derived multipotent mesenchymal stromal cells involved in placental angiogenesis via the PDGF-BB and STAT3 pathways. Biol Reprod 2015; 93:103. [PMID: 26353894 DOI: 10.1095/biolreprod.115.131250] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 09/08/2015] [Indexed: 12/23/2022] Open
Abstract
We studied the smooth muscle cell differentiation capability of human placental multipotent mesenchymal stromal cells (hPMSCs) and identified how endothelial cells recruit hPMSCs participating in vessel formation. hPMSCs from term placentas were induced to differentiate into smooth muscle cells under induction conditions and different matrix substrates. We assessed endothelial cells from umbilical veins for platelet-derived growth factor (PDGF)-BB expression and to induce hPMSC PDGFR-beta and STAT3 activation. Endothelial cells were co-cultured with hPMSCs for in vitro angiogenesis. Cell differentiation ability was then further assessed by mouse placenta transplantation assay. hPMSCs can differentiate into smooth muscle cells; collagen type I and IV or laminin support this differentiation. Endothelial cells expressed significant levels of PDGF-BB and activated STAT3 transcriptional activity in hPMSCs. Endothelial cell-conditioned medium induced hPMSC migration, which was inhibited by STAT3 small interfering RNA transfection or by pretreatement with PDGFR-beta-blocking antibody but not by PDGFR-alpha-blocking antibody or isotype immunoglobulin G (IgG; P < 0.001). hPMSCs can incorporate into endothelial cells with tube formation and promote endothelial cells, forming capillary-like networks than endothelial cells alone (tube lengths: 12 024.1 ± 960.1 vs. 9404.2 ± 584.7 pixels; P < 0.001). Capillary-like networks were significantly reduced by hPMSCs pretreated with PDGFR-beta-blocking antibody but not by PDGFR-alpha-blocking antibody or isotype IgG (P < 0.001). Transplantation of hPMSCs into mouse placentas revealed incorporation of the hPMSCs into vessel walls, which expressed alpha-smooth muscle actin, calponin, and smooth muscle myosin (heavy chain) in vivo. In conclusion, endothelial cell-hPMSC interactions occur during vessel development of placenta. Placental endothelial cell-derived PDGF-BB recruits hPMSCs involved in vascular development via PDGFR-beta/STAT3 activation.
Collapse
Affiliation(s)
- Cheng-Yi Chen
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Shu-Hsiang Liu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chia-Yu Chen
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Pei-Chun Chen
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chie-Pein Chen
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
17
|
Hemangioblastic foci in human first trimester placenta: Distribution and gestational profile. Placenta 2015; 36:1069-77. [PMID: 26318645 DOI: 10.1016/j.placenta.2015.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 08/01/2015] [Accepted: 08/10/2015] [Indexed: 11/20/2022]
Abstract
INTRODUCTION The human placenta is a site of both hematopoiesis and vasculogenesis. There are reports of hemangioblastic foci (HAF) in the first trimester placenta, but little published information about their spatiotemporal incidence. METHODS We have used semi-thin sections and whole mount staining techniques on archival early pregnancy hysterectomy material as well as freshly-collected termination tissue. RESULTS We report a description of the distribution of HAF, their gestational profile, and some characteristics of the constituent cells. We show crypt-shaped HAF are present in villi at different levels from 4 to 11 weeks and in the chorionic plate from 4 to 9 weeks. In the villous placenta, the foci often approach closely at one end to the trophoblast basement membrane. Morphologically they show remarkable similarity to those found in the yolk sac at similar stages. In some crypts, all cells are CD34+, but CD34 and nestin progressively segregate into the endothelial lineage. Brachyury is present in less differentiated cells. The erythroid lineage is dominant, as shown by the widespread expression of CD235a/glycophorin and characteristic erythroid morphologies, indicating various degrees of differentiation. However, CD41 is also present in non-endothelial cells. Initially a discontinuous UEA-1/CD31-positive endothelium forms at the periphery of the foci. These cells appear to become integrated into the developing vasculogenic/angiogenic vessel network. We also demonstrate that, independent of HAF, vasculogenesis occurs near the tips of growing villi during the first trimester. DISCUSSION We suggest HAF interface with the developing vascular network, producing communication channels that allow erythrocytes to enter the placental-embryonic circulation. We speculate that the erythroid cells act as oxygen reservoirs during the period before flow of maternal blood through the intervillous space of the placenta, allowing a slow feed of oxygen-rich cells to the developing embryo.
Collapse
|
18
|
Li J, Zeng G, Qi Y, Tang X, Zhang J, Wu Z, Liang J, Shi L, Liu H, Zhang P. Xenotransplantation of human adipose-derived stem cells in zebrafish embryos. PLoS One 2015; 10:e0123264. [PMID: 25849455 PMCID: PMC4388839 DOI: 10.1371/journal.pone.0123264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/17/2015] [Indexed: 12/18/2022] Open
Abstract
Zebrafish is a widely used animal model with well-characterized background in developmental biology. The fate of human adipose-derived stem cells (ADSCs) after their xenotransplantation into the developing embryos of zebrafish is unknown. Therefore, human ADSCs were firstly isolated, and then transduced with lentiviral vector system carrying a green fluorescent protein (GFP) reporter gene, and followed by detection of their cell viability and the expression of cell surface antigens. These GFP-expressing human ADSCs were transplanted into the zebrafish embryos at 3.3–4.3 hour post-fertilization (hpf). Green fluorescent signal, the proliferation and differentiation of human ADSCs in recipient embryos were respectively examined using fluorescent microscopy and immunohistochemical staining. The results indicated that human ADSCs did not change their cell viability and the expression levels of cell surface antigens after GFP transduction. Microscopic examination demonstrated that green fluorescent signals of GFP expressed in the transplanted cells were observed in the embryos and larva fish at post-transplantation. The positive staining of Ki-67 revealed the survival and proliferation of human ADSCs in fish larvae after transplantation. The expression of CD105 was observable in the xenotransplanted ADSCs, but CD31 expression was undetectable. Therefore, our results indicate that human ADSCs xenotransplanted in the zebrafish embryos not only can survive and proliferate at across-species circumstance, but also seem to maintain their undifferentiation status in a short term. This xenograft model of zebrafish embryos may provide a promising and useful technical platform for the investigation of biology and physiology of stem cells in vivo.
Collapse
Affiliation(s)
- Jin Li
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Guofang Zeng
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Yawei Qi
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Xudong Tang
- Institute of Biochemistry and Molecular Biology, Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Jingjing Zhang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Zeyong Wu
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Jie Liang
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Lei Shi
- School of Light Industry and Food Science, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Hongwei Liu
- Department of Plastic Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory for Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
- * E-mail: (HL); (PZ)
| | - Peihua Zhang
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
- * E-mail: (HL); (PZ)
| |
Collapse
|
19
|
Immunosuppressive therapy in allograft transplantation: from novel insights and strategies to tolerance and challenges. Cent Eur J Immunol 2014; 39:400-9. [PMID: 26155155 PMCID: PMC4440012 DOI: 10.5114/ceji.2014.45955] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 07/03/2014] [Indexed: 01/07/2023] Open
Abstract
Immunosuppression therapy is the key to successful post-transplantation outcomes. The need for ideal immunosuppression became durable maintenance of long-term graft survival. In spite of current immunosuppressive therapy regimens advances, surgical procedures, and preservation methods, organ transplantation is associated with a long-term poor survival and significant mortality. This has led to an increased interest to optimize outcomes while minimizing associated toxicity by using alternative methods for maintenance immunosuppression, organ rejection treatment, and monitoring of immunosuppression. T regulatory (Treg) cells, which have immunosuppressive functions and cytokine profiles, have been studied during the last decades. Treg cells are able to inhibit the development of allergen-specific cell responses and consequently play a key role in a healthy immune response to allergens. Mature dendritic cells (DCs) play a crucial role in the differentiation of Tregs, which are known to regulate allergic inflammatory responses. Advance in long-standing allograft outcomes may depend on new drugs with novel mechanisms of action with minimal toxicity. Newer treatment techniques have been developed, including using novel stem cell-based therapies such as mesenchymal stem cells, phagosomes and exosomes. Immunoisolation techniques and salvage therapies, including photopheresis and total lymphoid irradiation have emerged as alternative therapeutic choices. The present review evaluates the recent clinical advances in immunosuppressive therapies for organ transplantation.
Collapse
|
20
|
|
21
|
Heazlewood CF, Sherrell H, Ryan J, Atkinson K, Wells CA, Fisk NM. High incidence of contaminating maternal cell overgrowth in human placental mesenchymal stem/stromal cell cultures: a systematic review. Stem Cells Transl Med 2014; 3:1305-11. [PMID: 25154781 DOI: 10.5966/sctm.2014-0051] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Placenta is a readily accessible translationally advantageous source of mesenchymal stem/stromal cells (MSCs) currently used in cryobanking and clinical trials. MSCs cultured from human chorion have been widely assumed to be fetal in origin, despite evidence that placental MSCs may be contaminated with maternal cells, resulting in entirely maternally derived MSC cultures. To document the frequency and determinants of maternal cell contamination in chorionic MSCs, we undertook a PRISMA-compliant systematic review of publications in the PubMed, Medline, and Embase databases (January 2000 to July 2013) on placental and/or chorionic MSCs from uncomplicated pregnancies. Of 147 studies, only 26 (18%) investigated fetal and/or maternal cell origin. After excluding studies that did not satisfy minimal MSC criteria, 7 of 15 informative studies documented MSC cultures as entirely fetal, a further 7 studies reported cultured human chorionic MSC populations to be either maternal (n=6) or mixed (n=1), whereas 1 study separately cultured pure fetal and pure maternal MSC from the same placenta. Maternal cell contamination was associated with term and chorionic membrane samples and greater passage number but was still present in 30% of studies of chorionic villous MSCs. Although most studies assume fetal origin for MSCs sourced from chorion, this systematic review documents a high incidence of maternal-origin MSC populations in placental MSC cultures. Given that fetal MSCs have more primitive properties than adult MSCs, our findings have implications for clinical trials in which knowledge of donor and tissue source is pivotal. We recommend sensitive methods to quantitate the source and purity of placental MSCs.
Collapse
Affiliation(s)
- Celena F Heazlewood
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Helen Sherrell
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Ryan
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kerry Atkinson
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Christine A Wells
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nicholas M Fisk
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology at the Translational Research Institute, and Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia; Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
22
|
Passipieri JA, Kasai-Brunswick TH, Suhett G, Martins AB, Brasil GV, Campos DB, Rocha NN, Ramos IP, Mello DB, Rodrigues DC, Christie BB, Silva-Mendes BJ, Balduíno A, Sá RM, Lopes LM, Goldenberg RC, Campos de Carvalho AC, Carvalho AB. Improvement of cardiac function by placenta-derived mesenchymal stem cells does not require permanent engraftment and is independent of the insulin signaling pathway. Stem Cell Res Ther 2014; 5:102. [PMID: 25145631 PMCID: PMC4354978 DOI: 10.1186/scrt490] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/18/2014] [Accepted: 08/08/2014] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION The objective of this work was to evaluate the efficacy of placenta-derived mesenchymal stem cell (MSC) therapy in a mouse model of myocardial infarction (MI). Since MSCs can be obtained from two different regions of the human term placenta (chorionic plate or villi), cells obtained from both these regions were compared so that the best candidate for cell therapy could be selected. METHODS For the in vitro studies, chorionic plate MSCs (cp-MSCs) and chorionic villi MSCs (cv-MSCs) were extensively characterized for their genetic stability, clonogenic and differentiation potential, gene expression, and immunophenotype. For the in vivo studies, C57Bl/6 mice were submitted to MI and, after 21 days, received weekly intramyocardial injections of cp-MSCs for 3 weeks. Cells were also stably transduced with a viral construct expressing luciferase, under the control of the murine stem cell virus (MSCV) promoter, and were used in a bioluminescence assay. The expression of genes associated with the insulin signaling pathway was analyzed in the cardiac tissue from cp-MSCs and placebo groups. RESULTS Morphology, differentiation, immunophenotype, and proliferation were quite similar between these cells. However, cp-MSCs had a greater clonogenic potential and higher expression of genes related to cell cycle progression and genome stability. Therefore, we considered that the chorionic plate was preferable to the chorionic villi for the isolation of MSCs. Sixty days after MI, cell-treated mice had a significant increase in ejection fraction and a reduction in end-systolic volume. This improvement was not caused by a reduction in infarct size. In addition, tracking of cp-MSCs transduced with luciferase revealed that cells remained in the heart for 4 days after the first injection but that the survival period was reduced after the second and third injections. Quantitative reverse transcription-polymerase chain reaction revealed similar expression of genes involved in the insulin signaling pathway when comparing cell-treated and placebo groups. CONCLUSIONS Improvement of cardiac function by cp-MSCs did not require permanent engraftment and was not mediated by the insulin signaling pathway.
Collapse
Affiliation(s)
- Juliana A Passipieri
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Cardiologia, Rua das Laranjeiras 374, Rio de Janeiro, 22240-006, Brazil.
| | - Tais H Kasai-Brunswick
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Cardiologia, Rua das Laranjeiras 374, Rio de Janeiro, 22240-006, Brazil.
| | - Grazielle Suhett
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Rua Rodolpho Paulo Rocco 255, Rio de Janeiro, 21941-913, Brazil.
| | - Andreza B Martins
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Cardiologia, Rua das Laranjeiras 374, Rio de Janeiro, 22240-006, Brazil.
| | - Guilherme V Brasil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Dilza B Campos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Nazareth N Rocha
- Universidade Federal Fluminense, Rua Professor Hernani Melo 101, Niterói, 24210-130, Brazil.
| | - Isalira P Ramos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Rua Rodolpho Paulo Rocco 255, Rio de Janeiro, 21941-913, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Av Carlos Chagas Filho 373, Rio de Janeiro, 21941-902, Brazil.
| | - Debora B Mello
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Deivid C Rodrigues
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Beatriz B Christie
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Bernardo J Silva-Mendes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Alex Balduíno
- Centro de Pesquisa, Tecnologia e Inovação, Universidade Veiga de Almeida, Rua Ibituruna 108, Rio de Janeiro, 20271-020, Brazil.
| | - Renato M Sá
- Centro Pré-Natal de Diagnóstico e Tratamento, Clínica Perinatal, Rua das Laranjeiras 445, Rio de Janeiro, 22240-002, Brazil.
| | - Laudelino M Lopes
- Centro Pré-Natal de Diagnóstico e Tratamento, Clínica Perinatal, Rua das Laranjeiras 445, Rio de Janeiro, 22240-002, Brazil.
- Department of Obstetrics and Gynecology, Western University, London Health Sciences Centre-Victoria Hospital, B2-401, London, ON, N6H 5W9, Canada.
| | - Regina C Goldenberg
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Av Carlos Chagas Filho 373, Rio de Janeiro, 21941-902, Brazil.
| | - Antonio C Campos de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Cardiologia, Rua das Laranjeiras 374, Rio de Janeiro, 22240-006, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Av Carlos Chagas Filho 373, Rio de Janeiro, 21941-902, Brazil.
| | - Adriana B Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av Carlos Chagas Filho 373, Sala G2-053, Rio de Janeiro, RJ, 21941-902, Brazil.
- Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Av Carlos Chagas Filho 373, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
23
|
Chen CP, Chen YY, Huang JP, Wu YH. The effect of conditioned medium derived from human placental multipotent mesenchymal stromal cells on neutrophils: possible implications for placental infection. Mol Hum Reprod 2014; 20:1117-25. [PMID: 25140001 DOI: 10.1093/molehr/gau062] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The role of human placental multipotent mesenchymal stromal cells (hPMSCs) in placental inflammation is unknown. We hypothesize that hPMSCs are involved in the early phases of placental infection. hPMSCs were isolated from term placentas and neutrophils from peripheral blood. The expression of toll-like receptors (TLRs) and cytokines by hPMSCs was determined by RT-PCR, flow cytometry and enzyme-linked immunosorbent assay. The effect of conditioned medium of hPMSCs with or without lipopolysaccharide (LPS) pretreatment on neutrophil functions: migration, apoptosis and production of reactive oxygen species (ROS) was assessed by flow cytometry and western blot. hPMSCs expressed TLR1, TLR3, TLR4, TLR6, TLR7 and TLR9. LPS stimulation increased the expression of TLR4 and the production of IL-6 and IL-8 by hPMSCs. Neutrophils exhibited chemotaxis to hPMSC-conditioned medium, which was inhibited by IL-8 depletion. Neutrophil CD11b activation was promoted by hPMSC-conditioned medium, which was further enhanced in media from hPMSCs pretreated with LPS. hPMSC-conditioned medium reduced neutrophil ROS production. Neutrophil phagocytosis was increased by LPS alone but not by hPMSC-conditioned medium with or without LPS stimulation. hPMSC-conditioned medium induced STAT3 activation in neutrophils, which was inhibited by neutralizing antibody to IL-6. hPMSC-conditioned medium rescued neutrophils from apoptosis, but this effect was significantly reduced in conditioned medium of hPMSCs with LPS pretreatment. Depletion of IL-6 from the conditioned medium further inhibited the anti-apoptotic effect on neutrophils. Our results demonstrate that hPMSCs can interact with peripheral blood neutrophils in response to inflammatory signals of the placenta. Cytokines produced by hPMSCs can induce neutrophil chemotaxis and reduce neutrophil apoptosis.
Collapse
Affiliation(s)
- Chie-Pein Chen
- Division of High Risk Pregnancy, Mackay Memorial Hospital, 92 Sec. 2 Zhong-Shan North Road, Taipei 104, Taiwan Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yi-Yung Chen
- Division of High Risk Pregnancy, Mackay Memorial Hospital, 92 Sec. 2 Zhong-Shan North Road, Taipei 104, Taiwan
| | - Jian-Pei Huang
- Division of High Risk Pregnancy, Mackay Memorial Hospital, 92 Sec. 2 Zhong-Shan North Road, Taipei 104, Taiwan
| | - Yi-Hsin Wu
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
24
|
Youssef A, Iosef C, Han VKM. Low-oxygen tension and IGF-I promote proliferation and multipotency of placental mesenchymal stem cells (PMSCs) from different gestations via distinct signaling pathways. Endocrinology 2014; 155:1386-97. [PMID: 24506070 DOI: 10.1210/en.2013-1842] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The microenvironment of placental mesenchymal stem cells (PMSCs) is dynamic throughout gestation and determines changes in cell fate. In vivo, PMSCs initially develop in low-oxygen tension and low IGF-I concentrations, and both increase gradually with gestation. The impact of varying concentrations of IGF-I and changing oxygen tension on PMSC signaling and multipotency was investigated in PMSCs from early (preterm) and late (term) gestation human placentae. Preterm PMSCs had greater proliferative response to IGF-I, which was further enhanced by low-oxygen tension. Low-oxygen tension alone was sufficient to induce ERK1/2 phosphorylation, whereas IGF-I was required for AKT (protein kinase B) phosphorylation. Low-oxygen tension prolonged ERK1/2 and AKT phosphorylation with a slowed phosphorylation decay even in presence of IGF-I. Low-oxygen tension maintained higher levels of IGF-I receptor and insulin receptor substrate 1 that were otherwise decreased by exposure to IGF-I and induced a differential phosphorylation pattern on IGF-I receptorβ and insulin receptor substrate 1. Phosphorylation of ERK1/2 and AKT was different between the preterm and term PMSCs, and phospho-AKT, and not phospho-ERK1/2, was the major determinant of PMSC proliferation and octamer-4 levels. These studies demonstrate that low-oxygen tension regulates the fate of PMSCs from early and late gestations in response to IGF-I, both independently and dependently, via specific signal transduction mechanisms.
Collapse
Affiliation(s)
- Amer Youssef
- Departments of Biochemistry (A.Y., V.K.M.H.) and Paediatrics (C.I., V.K.M.H.), Schulich School of Medicine and Dentistry; Children's Health Research Institute (A.Y., C.I., V.K.M.H.); and Lawson Health Research Institute (A.Y., C.I., V.K.M.H.); Western University, London, Ontario, Canada N6C 2V5
| | | | | |
Collapse
|
25
|
Feasibility and potential of in utero foetal membrane-derived cell transplantation. Cell Tissue Bank 2013; 15:241-9. [DOI: 10.1007/s10561-013-9402-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 10/09/2013] [Indexed: 02/06/2023]
|
26
|
In utero hepatocellular transplantation in rats. Clin Dev Immunol 2013; 2013:562037. [PMID: 24062778 PMCID: PMC3766607 DOI: 10.1155/2013/562037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 06/14/2013] [Accepted: 06/25/2013] [Indexed: 12/28/2022]
Abstract
This work represents a step forward in the experimental design of an in utero hepatocellular transplantation model in rats. We focused on the enrichment optimization of isolated fetal hepatocytes suspension, arranging the surgery methodology of in utero transplantation, monitoring the biodistribution of the transplanted hepatocytes, and assessing the success of the transplants. Rat fetuses have been transplanted at the 17th embryonic day (ED17) with fetal hepatocytes isolated from rats at the end of pregnancy (ED21). We assessed possible differences between lymphocyte population, CD4 positive, CD8 positive, double-positive T-cells, and anti-inflammatory cytokines interleukins 4 and 10 (IL4 and IL10) as well. Cellular viability reached the rates of 90–95%. Transplanted groups had a limited success. Transplanted hepatocytes were not able to pass through the hematoplacental barrier. The hepatocytes injected were primarily located in the liver. There was an upward trend in the whole amount of T CD4 and T CD8 cells. There was an increased IL4 in the transplanted groups observed in the pregnant rats. The possibility to induce tolerance in fetuses with a hepatocyte transplant in utero could be a key point to avoid the immunosuppression treatments which must be undergone by transplanted patients.
Collapse
|
27
|
Makhoul G, Chiu RCJ, Cecere R. Placental mesenchymal stem cells: a unique source for cellular cardiomyoplasty. Ann Thorac Surg 2013; 95:1827-33. [PMID: 23541427 DOI: 10.1016/j.athoracsur.2012.11.053] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 11/15/2012] [Accepted: 11/16/2012] [Indexed: 02/06/2023]
Abstract
In coronary heart disease, the use of stem cells for regeneration purposes has been broadly studied. Whereas bone marrow mesenchymal stem cells remain the most extensively investigated, other cell sources have been reported. Here we discuss and compare the characteristics of placenta-derived mesenchymal stem cells as a novel alternative cell source for cellular cardiomyoplasty. These cells are isolated from the human term placenta, which is normally discarded post partum. With their lack of ethical conflicts and young age, the readily available placenta-derived mesenchymal stem cells could be more suitable for myocardial regenerative therapy.
Collapse
Affiliation(s)
- Georges Makhoul
- Divisions of Cardiac and Experimental Surgery, McGill University Health Center, Montreal, Quebec, Canada
| | | | | |
Collapse
|
28
|
Yang CM, Gong XL, Qiu J, Tang HX, Gong ZJ, Huang SZ, Zeng F. Engraftment of genetically modified human amniotic fluid-derived progenitor cells to produce coagulation factor IX after in utero transplantation in mice. Cell Biol Int 2013; 37:420-9. [PMID: 23504762 DOI: 10.1002/cbin.10037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 12/23/2012] [Indexed: 12/29/2022]
Abstract
Human amniotic fluid derived progenitor cells (hAFPCs) may be multipotent and can be considered a potential tool in the field of cell therapy for haemophilia B. Their capacity to express human coagulation factor IX (hFIX) after transduction and their fate after in utero transplantation is unknown. hAFPCs isolated from second trimester pregnancies were assessed for their phenotypic markers, multilineage capacity, and expression of hFIX after transduction. Their engraftment potential was analysed in a mouse model after in utero transplantation at embryonic day 12.5. Immunohistochemistry, fluorescence in situ, ELISA and PCR were used to assess post-transplant chimeras. hAFPCs expressed several pluripotent markers, including NANOG, SOX2, SSEA4 and TRA-1-60, and could differentiate into adipocytes and osteocytes. In vitro, after transduction with hFIX and EGFP cDNAs, constitutive hFIX protein expression and clotting activity were found. Engraftment was achieved in various foetal tissues after in utero transplantation. Safe engraftment without oncogenesis was confirmed, with low donor cell levels, but persistent engraftment, into different organs (liver, heart and lung) through to 12 weeks of age. Transgenic expression of circulating hFIX was detected in recipient mice for up to 12 weeks. hAFPCs can be engrafted long-term in immunocompetent mice after in utero transplantation. Thus, cell transplantation approaches using genetically engineered hAFPCs may prove valuable for the prenatal treatment for haemophilia B.
Collapse
Affiliation(s)
- Chen-Min Yang
- Laboratory of Development Biology, Institute of Medical Science, Shanghai Jiao Tong University School of Medicine, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Pipino C, Shangaris P, Resca E, Zia S, Deprest J, Sebire NJ, David AL, Guillot PV, De Coppi P. Placenta as a reservoir of stem cells: an underutilized resource? Br Med Bull 2013. [PMID: 23184854 DOI: 10.1093/bmb/lds033] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Both embryonic and adult tissues are sources of stem cells with therapeutic potential but with some limitations in the clinical practice such as ethical considerations, difficulty in obtaining and tumorigenicity. As an alternative, the placenta is a foetal tissue that can be obtained during gestation and at term, and it represents a reservoir of stem cells with various potential. SOURCES OF DATA We reviewed the relevant literature concerning the main stem cells that populate the placenta. AREAS OF AGREEMENT Recently, the placenta has become useful source of stem cells that offer advantages in terms of proliferation and plasticity when compared with adult cells and permit to overcome the ethical and safety concern inherent in embryonic stem cells. In addition, the placenta has the advantage of containing epithelia, haematopoietic and mesenchymal stem cells. These stem cells possess immunosuppressive properties and have the capacity of suppress in vivo inflammatory responses. AREAS OF CONTROVERSY Some studies describe a subpopulation of placenta stem cells expressing pluripotency markers, but for other studies, it is not clear whether pluripotent stem cells are present during gestation beyond the first few weeks. Particularly, the expression of some pluripotency markers such as SSEA-3, TRA-1-60 and TRA-1-81 has been reported by us, but not by others. GROWING POINTS Placenta stem cells could be of great importance after delivery for banking for autologous and allogeneic applications. The beneficial effects of these cells may be due to secretion of bioactive molecules that act through paracrine actions promoting beneficial effects. AREAS TIMELY FOR DEVELOPING RESEARCH Understanding the role of placenta stem cells during pregnancy and their paracrine actions could help in the study of some diseases that affect the placenta during pregnancy.
Collapse
|
30
|
Choi SA, Choi HS, Kim KJ, Lee DS, Lee JH, Park JY, Kim EY, Li X, Oh HY, Lee DS, Kim MK. Isolation of canine mesenchymal stem cells from amniotic fluid and differentiation into hepatocyte-like cells. In Vitro Cell Dev Biol Anim 2012; 49:42-51. [PMID: 23242927 DOI: 10.1007/s11626-012-9569-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 11/05/2012] [Indexed: 11/24/2022]
Abstract
Recent findings have demonstrated that amniotic fluid cells are an interesting and potential source of mesenchymal stem cells (MSCs). In this study, we isolated MSCs from canine amniotic fluid and then characterized their multilineage differentiation ability. Canine amniotic fluid stem (cAFS) cells at passage 5 had a fibroblast-like morphology instead of forming colonies and were positive for pluripotent stem cell markers such as OCT4, NANOG, and SOX2. Flow cytometry analysis showed the expression of MSC surface markers CD44, CD29, and CD90 on the cAFS cells. In addition, these cells were cultured under conditions favorable for adipogenic, chondrogenic, and osteogenic induction. The results of this experiment confirmed the mesenchymal nature of cAFS cells and their multipotent potential. Interestingly, although the cells exhibited a fibroblast-like morphology after hepatogenic induction, reverse transcription-polymerase chain reaction revealed that the expression of several hepatic genes, such as albumin, tyrosine aminotransferase, and alpha-1 antiproteinase, increased following maturation and differentiation. These findings indicated that cAFS cells have functional properties similar to those of hepatocytes. Taken together, the results of our study demonstrated that cAFS cells with mesenchymal characteristics can be successfully isolated from canine amniotic fluid and possess functional properties characteristic of hepatocytes. The findings of our work suggest that cAFS cells have the potential to be a resource for cell-based therapies in a canine model of hepatic disease.
Collapse
Affiliation(s)
- Seon-A Choi
- Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Daejeon, 305-764, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Vosdoganes P, Lim R, Moss TJM, Wallace EM. Cell therapy: a novel treatment approach for bronchopulmonary dysplasia. Pediatrics 2012; 130:727-37. [PMID: 22945412 DOI: 10.1542/peds.2011-2576] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a major cause of substantial lifelong morbidity in preterm infants. Despite a better understanding of the pathophysiology of BPD and significant research effort into its management, there remains today no effective treatment. Cell-based therapy is a novel approach that offers much promise in the prevention and treatment of BPD. Recent research supports a therapeutic role for cell transplantation in the management of a variety of acute and chronic adult and childhood lung diseases, with potential of such therapy to reduce inflammation and prevent acute lung injury. However, considerable uncertainties remain regarding cell therapies before they can be established as safe and effective clinical treatments for BPD. This review summarizes the current literature investigating cell therapies in lung disease, with particular focus on the various types of cells available and their specific properties in the context of a future therapy for BPD.
Collapse
Affiliation(s)
- Patricia Vosdoganes
- Ritchie Centre, Department of Obstetrics and Gynaecology, Monash Medical Centre, 246 Clayton Rd, Clayton, VIC, Australia 3168
| | | | | | | |
Collapse
|
32
|
Abumaree M, Al Jumah M, Pace RA, Kalionis B. Immunosuppressive properties of mesenchymal stem cells. Stem Cell Rev Rep 2012; 8:375-92. [PMID: 21892603 DOI: 10.1007/s12015-011-9312-0] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSC) can be isolated from different adult tissues including bone marrow, adipose tissue, cord blood and placenta. MSCs modulate the immune function of the major immune cell populations involved in alloantigen recognition and elimination, including antigen presenting cells, T cells, B cells and natural killer cells. Many clinical trials are currently underway that employ MSCs to treat human immunological diseases. However, the molecular mechanism that mediates the immunosuppressive effect of MSCs is still unclear and the safety of using MSC in patient needs further confirmation. Here, we review the cytokines that activate MSCs and the soluble factors produced by MSCs, which allow them to exert their immunosuppressive effects. We review the mechanism responsible, at least in part, for the immune suppressive effects of MSCs and highlight areas of research required for a better understanding of MSC immune modulation.
Collapse
Affiliation(s)
- Mohamed Abumaree
- King Saud Bin Abdulaziz University for Health Sciences/King Abdullah International Medical Research Center, King Abdulaziz Medical City, National Guard Health Affairs, PO Box 22490, Riyadh, 11426, Mail Code 1515, Saudi Arabia
| | | | | | | |
Collapse
|
33
|
Martínez-González I, Moreno R, Petriz J, Gratacós E, Aran JM. Engraftment potential of adipose tissue-derived human mesenchymal stem cells after transplantation in the fetal rabbit. Stem Cells Dev 2012; 21:3270-7. [PMID: 22738094 DOI: 10.1089/scd.2012.0032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Due to their favorable intrinsic features, including engraftment, differentiation, and immunomodulatory potential, adult mesenchymal stem cells (MSCs) have been proposed for therapeutic in utero intervention. Further improvement of such attributes for particular diseases might merely be achieved by ex vivo MSC genetic engineering previous to transplantation. Here, we evaluated for the first time the feasibility, biodistribution, long-term engraftment, and transgenic enhanced green fluorescent protein (EGFP) expression of genetically engineered human adipose tissue-derived MSCs (EGFP(+)-ASCs) after intra-amniotic xenotransplantation at E17 of gestation into our validated pregnant rabbit model. Overall, the procedure was safe (86.4% survival rate; absence of anatomical defects). Stable, low-level engraftment of EGFP(+)-ASCs was confirmed by assessing the presence of the pWT-EGFP lentiviral provirus in the young transplanted rabbit tissues. Accordingly, similar frequencies of provirus-positive animals were found at both 8 weeks (60%) and 16 weeks (66.7%) after in utero intervention. The presence of EGFP(+)-ASCs was more frequent in respiratory epithelia (lung and trachea), according to the route of administration. However, we were unable to detect EGFP expression, neither by real-time polymerase chain reaction nor by immunohistochemistry, in the provirus-positive tissues, suggesting EGFP transgene silencing mediated by epigenetic events. Moreover, we noticed lack of both host cellular immune responses against xenogeneic ASCs and humoral immune responses against transgenic EGFP. Therefore, the fetal microchimerism achieved by the EGFP(+)-ASCs in the young rabbit hosts indicates induction of donor-specific tolerance after fetal rabbit xenotransplantation, which should boost postnatal transplantation for the early treatment/prevention of many devastating congenital disorders.
Collapse
Affiliation(s)
- Itziar Martínez-González
- Human Molecular Genetics Group, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | |
Collapse
|
34
|
The immunomodulatory and neuroprotective effects of mesenchymal stem cells (MSCs) in experimental autoimmune encephalomyelitis (EAE): a model of multiple sclerosis (MS). Int J Mol Sci 2012; 13:9298-9331. [PMID: 22942767 PMCID: PMC3430298 DOI: 10.3390/ijms13079298] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/11/2012] [Accepted: 07/11/2012] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that differentiate into the mesenchymal lineages of adipocytes, osteocytes and chondrocytes. MSCs can also transdifferentiate and thereby cross lineage barriers, differentiating for example into neurons under certain experimental conditions. MSCs have anti-proliferative, anti-inflammatory and anti-apoptotic effects on neurons. Therefore, MSCs were tested in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), for their effectiveness in modulating the pathogenic process in EAE to develop effective therapies for MS. The data in the literature have shown that MSCs can inhibit the functions of autoreactive T cells in EAE and that this immunomodulation can be neuroprotective. In addition, MSCs can rescue neural cells via a mechanism that is mediated by soluble factors, which provide a suitable environment for neuron regeneration, remyelination and cerebral blood flow improvement. In this review, we discuss the effectiveness of MSCs in modulating the immunopathogenic process and in providing neuroprotection in EAE.
Collapse
|
35
|
Kim J, Park S, Kang HM, Ahn CW, Kwon HC, Song JH, Lee YJ, Lee KH, Yang H, Baek SY, Yoo SH, Kim SH, Kim H. Human insulin secreted from insulinogenic xenograft restores normoglycemia in type 1 diabetic mice without immunosuppression. Cell Transplant 2012; 21:2131-47. [PMID: 22490341 DOI: 10.3727/096368912x636803] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In the present study, we examined the therapeutic potential of human amnion-derived insulin-secreting cells for type 1 diabetes. Human amniotic mesenchymal stem cells (hAMs) were isolated from amnion and cultivated to differentiate into insulin-secreting cells in vitro. After culture in vitro, the differentiated cells (hAM-ISCs) were intensively stained with dithizone and secreted insulin and c-peptide in a high-glucose-dependent manner. They expressed mRNAs of pancreatic cell-related genes, including INS, PDX1, Nkx6-1, NEUROG3, ISL1, NEUROD1, GLUT1, GLUT2, PC1/3, PC2, GCK, PPY, SST, and GC, and were positive for human insulin and c-peptide. Transplantation of hAM-ISCs into the kidneys of mice with streptozotocin-induced diabetes restored body weight and normalized the blood glucose levels, which lasted for 210 days. Only human insulin and c-peptide were detected in the blood of normalized mice after 2 months of transplantation, but little mouse insulin and c-peptide. Removal of graft-bearing kidneys from these mice resulted in causing hyperglycemia again. Human cell-specific gene, hAlu, and human pancreatic cell-specific genes, insulin, PDX1, GLUT1, GLP1R, Nkx6-1, NEUROD1, and NEUROG3, were detected in the graft-bearing kidneys. Colocalization of human insulin and human nuclei antigen was also observed. These results demonstrate that hAMs could differentiate into functional insulin-secreting cells in vitro, and human insulin secreted from hAM-ISCs following transplantation into type 1 diabetic mice could normalize hyperglycemia, overcoming immune rejection for a long period.
Collapse
Affiliation(s)
- J Kim
- bcellbio, Inc., Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
This study aims to modify a cyclodextrin-PEI-based polymer, PEI-β-CyD, with the TAT peptide for plasmid DNA delivery to placenta mesenchymal stem cells (PMSCs). By using the disulfide exchange between the SPDP-activated PEI-β-CyD and TAT peptide, the TAT-PEI-β-CyD polymer was fabricated and the success of this was confirmed by the presence of characteristic peaks for PEI (at δ 2.8-3.2 ppm), CyD (at δ 5.2, 3.8-4.0 and 3.4-3.6 ppm) and TAT (at δ 1.6-1.9 and 6.8-7.2 ppm) in the (1)H NMR spectrum of TAT-PEI-β-CyD. The polymer-plasmid-DNA polyplex could condense DNA at an N/P ratio of 7.0-8.0, and form nanoparticles with the size of 150.6±5.6 nm at its optimal N/P ratio (20/1). By examining the transfection efficiency and cytotoxicity of TAT-PEI-β-CyD, conjugation of the TAT peptide onto PEI-β-CyD was demonstrated to improve the transfection efficiency of PEI-β-CyD in PMSCs after 48 and 96 hours of post-transfection incubation. The viability of PEI-β-CyD-treated PMSCs was shown to be over 80% after 5 h of treatment and 24 h of post-treatment incubation. In summary, this study showed that the TAT-PEI-β-CyD polymer as a vector for plasmid DNA delivery to PMSCs and other cells warrants further investigations. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s12668-011-0010-9) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
Moreno R, Martínez-González I, Rosal M, Nadal M, Petriz J, Gratacós E, Aran JM. Fetal liver-derived mesenchymal stem cell engraftment after allogeneic in utero transplantation into rabbits. Stem Cells Dev 2011; 21:284-95. [PMID: 21495909 DOI: 10.1089/scd.2010.0483] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Prenatal transplantation of genetically engineered mesenchymal stem cells (MSCs) might benefit prevention or treatment of early-onset genetic disorders due to the cells' intrinsic regenerative potential plus the acquired advantage from therapeutic transgene expression. However, a thorough assessment of the safety, accessibility, and behavior of these MSCs in the fetal environment using appropriate animal models is required before we can advance toward a clinical application. We have recently shown that fetal rabbit liver MSCs (fl-MSCs) have superior growth rate, clonogenic capability, and in vitro adherence and differentiation abilities compared with adult rabbit bone marrow MSCs. In this follow-up study, we report safe and widespread distribution of recombinant pSF-EGFP retrovirus-transduced fl-MSCs (EGFP(+)-fl-MSCs) in neonatal rabbit tissues at 10 days after fetal allogeneic transplantation through both intrahepatic and intra-amniotic administration. Conversely, a more restricted biodistribution pattern according to the route of administration was apparent in the young rabbits intervened at 16 weeks after fetal EGFP(+)-fl-MSC transplantation. Furthermore, the presence of these cells in the recipients' tissues, tracked with the reporter provirus, was inversely related to the developmental stage of the fetuses at the time of intervention. Long-term engraftment was confirmed both by fluorescence in situ hybridization analysis on touch tissue imprints using a chromosome Y-specific BAC probe, and by immunohistochemical localization of EGFP expression. Finally, there was no evidence of immune responses against the transplanted EGFP(+)-fl-MSCs or the EGFP transgenic product in the treated young rabbits. Thus, cell transplantation approaches using genetically engineered fetal MSCs may prove particularly valuable to frontier medical treatments for congenital birth defects in perinatology.
Collapse
Affiliation(s)
- Rafael Moreno
- Human Molecular Genetics Group, Institut d’Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
38
|
Parolini O, Caruso M. Review: Preclinical studies on placenta-derived cells and amniotic membrane: an update. Placenta 2011; 32 Suppl 2:S186-95. [PMID: 21251712 DOI: 10.1016/j.placenta.2010.12.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 12/10/2010] [Accepted: 12/13/2010] [Indexed: 02/06/2023]
Abstract
Recent years have seen considerable advances in our knowledge of the biology and properties of stem/progenitor cells isolated from placental tissues. This has encouraged researchers to address the potential effects of these cells in animal models of different diseases, resulting in increasing expectations regarding their possible utility for cell-based therapeutic applications. This rapidly evolving research field is also enriched by studies aimed at expanding the use of the whole amniotic membrane (AM), a well-known surgical material, for pathological conditions other than those tested so far and for which clinical applications already exist. In this review, we provide an update on studies that have been performed with placenta-derived cells and fragments of the entire AM to validate their potential clinical applications in a variety of diseases, in particular those associated with degenerative processes induced by inflammatory and fibrotic mechanisms. We also offer, as far as possible, insight into the interpretation and suggested mechanisms to explain the most important outcomes achieved to date.
Collapse
Affiliation(s)
- O Parolini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Brescia, Italy.
| | | |
Collapse
|
39
|
Bieback K, Brinkmann I. Mesenchymal stromal cells from human perinatal tissues: From biology to cell therapy. World J Stem Cells 2010; 2:81-92. [PMID: 21607124 PMCID: PMC3097927 DOI: 10.4252/wjsc.v2.i4.81] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 08/11/2010] [Accepted: 08/16/2010] [Indexed: 02/06/2023] Open
Abstract
Cell-based regenerative medicine is of growing interest in biomedical research. The role of stem cells in this context is under intense scrutiny and may help to define principles of organ regeneration and develop innovative therapeutics for organ failure. Utilizing stem and progenitor cells for organ replacement has been conducted for many years when performing hematopoietic stem cell transplantation. Since the first successful transplantation of umbilical cord blood to treat hematological malignancies, non-hematopoietic stem and progenitor cell populations have recently been identified within umbilical cord blood and other perinatal and fetal tissues. A cell population entitled mesenchymal stromal cells (MSCs) emerged as one of the most intensely studied as it subsumes a variety of capacities: MSCs can differentiate into various subtypes of the mesodermal lineage, they secrete a large array of trophic factors suitable of recruiting endogenous repair processes and they are immunomodulatory.Focusing on perinatal tissues to isolate MSCs, we will discuss some of the challenges associated with these cell types concentrating on concepts of isolation and expansion, the comparison with cells derived from other tissue sources, regarding phenotype and differentiation capacity and finally their therapeutic potential.
Collapse
Affiliation(s)
- Karen Bieback
- Karen Bieback, Irena Brinkmann, Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, DRK-Blutspendedienst Baden-Württemberg - Hessen gGmbH, Ludolf-Krehl-Str. 13-17, D-68167 Mannheim, Germany
| | | |
Collapse
|
40
|
Parolini O, Alviano F, Bergwerf I, Boraschi D, De Bari C, De Waele P, Dominici M, Evangelista M, Falk W, Hennerbichler S, Hess DC, Lanzoni G, Liu B, Marongiu F, McGuckin C, Mohr S, Nolli ML, Ofir R, Ponsaerts P, Romagnoli L, Solomon A, Soncini M, Strom S, Surbek D, Venkatachalam S, Wolbank S, Zeisberger S, Zeitlin A, Zisch A, Borlongan CV. Toward cell therapy using placenta-derived cells: disease mechanisms, cell biology, preclinical studies, and regulatory aspects at the round table. Stem Cells Dev 2010; 19:143-54. [PMID: 19947828 DOI: 10.1089/scd.2009.0404] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Among the many cell types that may prove useful to regenerative medicine, mounting evidence suggests that human term placenta-derived cells will join the list of significant contributors. In making new cell therapy-based strategies a clinical reality, it is fundamental that no a priori claims are made regarding which cell source is preferable for a particular therapeutic application. Rather, ongoing comparisons of the potentiality and characteristics of cells from different sources should be made to promote constant improvement in cell therapies, and such comparisons will likely show that individually tailored cells can address disease-specific clinical needs. The principle underlying such an approach is resistance to the notion that comprehensive characterization of any cell type has been achieved, neither in terms of phenotype nor risks-to-benefits ratio. Tailoring cell therapy approaches to specific conditions also requires an understanding of basic disease mechanisms and close collaboration between translational researchers and clinicians, to identify current needs and shortcomings in existing treatments. To this end, the international workshop entitled "Placenta-derived stem cells for treatment of inflammatory diseases: moving toward clinical application" was held in Brescia, Italy, in March 2009, and aimed to harness an understanding of basic inflammatory mechanisms inherent in human diseases with updated findings regarding biological and therapeutic properties of human placenta-derived cells, with particular emphasis on their potential for treating inflammatory diseases. Finally, steps required to allow their future clinical application according to regulatory aspects including good manufacturing practice (GMP) were also considered. In September 2009, the International Placenta Stem Cell Society (IPLASS) was founded to help strengthen the research network in this field.
Collapse
|
41
|
Abstract
Mesenchymal stem cells (MSCs) have great potential for treating various diseases, especially those related to tissue damage involving immune reactions. Various studies have demonstrated that MSCs are strongly immunosuppressive in vitro and in vivo. Our recent studies have shown that un-stimulated MSCs are indeed incapable of immunosuppression; they become potently immunosuppressive upon stimulation with the supernatant of activated lymphocytes, or with combinations of IFN-gamma with TNF-alpha, IL-1alpha or IL-1beta. This observation revealed that under certain circumstances, inflammatory cytokines can actually become immunosuppressive. We showed that there is a species variation in the mechanisms of MSC-mediated immunosuppression: immunosuppression by cytokine-primed mouse MSCs is mediated by nitric oxide (NO), whereas immunosuppression by cytokine-primed human MSCs is executed through indoleamine 2, 3-dioxygenase (IDO). Additionally, upon stimulation with the inflammatory cytokines, both mouse and human MSCs secrete several leukocyte chemokines that apparently serve to attract immune cells into the proximity with MSCs, where NO or IDO is predicted to be most active. Therefore, immunosuppression by inflammatory cytokine-stimulated MSCs occurs via the concerted action of chemokines and immune-inhibitory NO or IDO produced by MSCs. Thus, our results provide novel information about the mechanisms of MSC-mediated immunosuppression and for better application of MSCs in treating tissue injuries induced by immune responses.
Collapse
|
42
|
Angiogenesis in differentiated placental multipotent mesenchymal stromal cells is dependent on integrin alpha5beta1. PLoS One 2009; 4:e6913. [PMID: 19847290 PMCID: PMC2760707 DOI: 10.1371/journal.pone.0006913] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 07/30/2009] [Indexed: 12/20/2022] Open
Abstract
Human placental multipotent mesenchymal stromal cells (hPMSCs) can be isolated from term placenta, but their angiogenic ability and the regulatory pathways involved are not known. hPMSCs were shown to express integrins αv, α4, α5, β1, β3, and β5 and could be induced to differentiate into cells expressing endothelial markers. Increases in cell surface integrins α5 and β1, but not α4, αvβ3, or αvβ5, accompanied endothelial differentiation. Vascular endothelial growth factor-A augmented the effect of fibronectin in enhancing adhesion and migration of differentiated hPMSC through integrin α5β1, but not αvβ3 or αvβ5. Formation of capillary-like structures in vitro from differentiated cells was inhibited by pre-treatment with function-blocking antibodies to integrins α5 and β1. When hPMSCs were seeded onto chick chorioallantoic membranes (CAM), human von Willebrand factor-positive cells were observed to engraft in the chick endothelium. CAMs transplanted with differentiated hPMSCs had a greater number of vessels containing human cells and more incorporated cells per vessel compared to CAMs transplanted with undifferentiated hPMSCs, and overall angiogenesis was enhanced more by the differentiated cells. Function-blocking antibodies to integrins α5 and β1 inhibited angiogenesis in the CAM assay. These results suggest that differentiated hPMSCs may contribute to blood vessel formation, and this activity depends on integrin α5β1.
Collapse
|
43
|
Cargnoni A, Di Marcello M, Campagnol M, Nassuato C, Albertini A, Parolini O. Amniotic membrane patching promotes ischemic rat heart repair. Cell Transplant 2009; 18:1147-59. [PMID: 19650976 DOI: 10.3727/096368909x12483162196764] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The amniotic membrane has long been applied for wound healing and treatment of ophthalmological disorders, even though the mechanisms underlying its actions remain to be clarified. Recently, cells derived from fetal membranes of human term placenta have raised strong interest in regenerative medicine for their stem cell potential and immunomodulatory features. Our study aimed to investigate the possible utility of amniotic membrane to limit postischemic cardiac injury. A fragment of human amniotic membrane was applied onto the left ventricle of rats that had undergone ischemia through left anterior descending coronary artery ligation. Echocardiographic assessment of morphological and functional cardiac parameters was then performed over a 3-month period. We demonstrated that application of an amniotic membrane fragment onto ischemic rat hearts could significantly reduce postischemic cardiac dysfunction. The amniotic membrane-treated rats showed higher preservation of cardiac dimensions and improved cardiac contractile function in terms of higher left ventricle ejection fraction, fractional shortening, and wall thickening. These improvements were apparent by day 7 after application of the amniotic membrane, persisted for at least 2 months, and occurred independently of cardiac injury severity. No engraftment of amniotic cells was detected into host cardiac tissues. Our results suggest that use of amniotic membrane may constitute a convenient vehicle for supplying cells that produce cardioprotective soluble factors, and reinforce the notion that this tissue constitutes a cell source with clinical potential that has yet to be completely revealed.
Collapse
Affiliation(s)
- Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, 25124 Brescia, Italy
| | | | | | | | | | | |
Collapse
|