1
|
Abstract
Embryonic development and cell specification have been viewed as an epigenetically rigid process. Through accumulation of irreversible epigenetic marks, the differentiation process has been considered unidirectional, and once completed cell specification would be permanent and stable. However, somatic cell nuclear transfer that involved the implantation of a somatic nucleus into a previously enucleated oocyte accomplished in amphibians in the 1950s and in mammals in the late 1990s-resulting in the birth of "Dolly the sheep"-clearly showed that "terminal" differentiation is reversible. In parallel, work on lineage-determining factors like MyoD revealed surprising potential to modulate lineage identity in somatic cells. This work culminated in the discovery that a set of four defined factors can reprogram fibroblasts into induced pluripotent stem (iPS) cells, which were shown to be molecularly and functionally equivalent to blastocyst-derived embryonic stem (ES) cells, thus essentially showing that defined factors can induce authentic reprogramming without the need of oocytes. This concept was further extended when it was shown that fibroblasts can be directly converted into neurons, showing induced lineage conversion is possible even between cells representing two different germ layers. These findings suggest that "everything is possible" (i.e., once key lineage reprogramming factors are identified, cells should be able to convert into any desired lineage).
Collapse
Affiliation(s)
- Hannah Shelby
- Departments of Pathology and Chemical and Systems Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Tara Shelby
- Departments of Pathology and Chemical and Systems Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Marius Wernig
- Departments of Pathology and Chemical and Systems Biology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
2
|
Grath A, Dai G. Direct cell reprogramming for tissue engineering and regenerative medicine. J Biol Eng 2019; 13:14. [PMID: 30805026 PMCID: PMC6373087 DOI: 10.1186/s13036-019-0144-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/27/2019] [Indexed: 02/06/2023] Open
Abstract
Direct cell reprogramming, also called transdifferentiation, allows for the reprogramming of one somatic cell type directly into another, without the need to transition through an induced pluripotent state. Thus, it is an attractive approach to develop novel tissue engineering applications to treat diseases and injuries where there is a shortage of proliferating cells for tissue repair. In certain tissue damage, terminally differentiated somatic cells lose their ability to proliferate, as a result, damaged tissues cannot heal by themselves. Examples of these scenarios include myocardial infarctions, neurodegenerative diseases, and cartilage injuries. Transdifferentiation is capable of reprogramming cells that are abundant in the body into desired cell phenotypes that are able to restore tissue function in damaged areas. Therefore, direct cell reprogramming is a promising direction in the cell and tissue engineering and regenerative medicine fields. In recent years, several methods for transdifferentiation have been developed, ranging from the overexpression of transcription factors via viral vectors, to small molecules, to clustered regularly interspaced short palindromic repeats (CRISPR) and its associated protein (Cas9) for both genetic and epigenetic reprogramming. Overexpressing transcription factors by use of a lentivirus is currently the most prevalent technique, however it lacks high reprogramming efficiencies and can pose problems when transitioning to human subjects and clinical trials. CRISPR/Cas9, fused with proteins that modulate transcription, has been shown to improve efficiencies greatly. Transdifferentiation has successfully generated many cell phenotypes, including endothelial cells, skeletal myocytes, neuronal cells, and more. These cells have been shown to emulate mature adult cells such that they are able to mimic major functions, and some are capable of promoting regeneration of damaged tissue in vivo. While transdifferentiated cells have not yet seen clinical use, they have had promise in mice models, showing success in treating liver disease and several brain-related diseases, while also being utilized as a cell source for tissue engineered vascular grafts to treat damaged blood vessels. Recently, localized transdifferentiated cells have been generated in situ, allowing for treatments without invasive surgeries and more complete transdifferentiation. In this review, we summarized the recent development in various cell reprogramming techniques, their applications in converting various somatic cells, their uses in tissue regeneration, and the challenges of transitioning to a clinical setting, accompanied with potential solutions.
Collapse
Affiliation(s)
- Alexander Grath
- Department of Bioengineering, Northeastern University, Lake Hall 214A, 360 Huntington Avenue, Boston, MA 02115 USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Lake Hall 214A, 360 Huntington Avenue, Boston, MA 02115 USA
| |
Collapse
|
3
|
Wei ZJ, Fan BY, Liu Y, Ding H, Tang HS, Pan DY, Shi JX, Zheng PY, Shi HY, Wu H, Li A, Feng SQ. MicroRNA changes of bone marrow-derived mesenchymal stem cells differentiated into neuronal-like cells by Schwann cell-conditioned medium. Neural Regen Res 2019; 14:1462-1469. [PMID: 30964074 PMCID: PMC6524508 DOI: 10.4103/1673-5374.253532] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells differentiate into neurons under the induction of Schwann cells. However, key microRNAs and related pathways for differentiation remain unclear. This study screened and identified differentially expressed microRNAs in bone marrow-derived mesenchymal stem cells induced by Schwann cell-conditioned medium, and explored targets and related pathways involved in their differentiation into neuronal-like cells. Primary bone marrow-derived mesenchymal stem cells were isolated from femoral and tibial bones, while primary Schwann cells were isolated from bilateral saphenous nerves. Bone marrow-derived mesenchymal stem cells were cultured in unconditioned (control group) and Schwann cell-conditioned medium (bone marrow-derived mesenchymal stem cell + Schwann cell group). Neuronal differentiation of bone marrow-derived mesenchymal stem cells induced by Schwann cell-conditioned medium was observed by time-lapse imaging. Upon induction, the morphology of bone marrow-derived mesenchymal stem cells changed into a neural shape with neurites. Results of quantitative reverse transcription-polymerase chain reaction revealed that nestin mRNA expression was upregulated from 1 to 3 days and downregulated from 3 to 7 days in the bone marrow-derived mesenchymal stem cell + Schwann cell group. Compared with the control group, microtubule-associated protein 2 mRNA expression gradually increased from 1 to 7 days in the bone marrow-derived mesenchymal stem cell + Schwann cell group. After 7 days of induction, microRNA analysis identified 83 significantly differentially expressed microRNAs between the two groups. Gene Ontology analysis indicated enrichment of microRNA target genes for neuronal projection development, regulation of axonogenesis, and positive regulation of cell proliferation. Kyoto Encyclopedia of Genes and Genomes pathway analysis demonstrated that Hippo, Wnt, transforming growth factor-beta, and Hedgehog signaling pathways were potentially associated with neural differentiation of bone marrow-derived mesenchymal stem cells. This study, which carried out successful microRNA analysis of neuronal-like cells differentiated from bone marrow-derived mesenchymal stem cells by Schwann cell induction, revealed key microRNAs and pathways involved in neural differentiation of bone marrow-derived mesenchymal stem cells. All protocols were approved by the Animal Ethics Committee of Institute of Radiation Medicine, Chinese Academy of Medical Sciences on March 12, 2017 (approval number: DWLI-20170311).
Collapse
Affiliation(s)
- Zhi-Jian Wei
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bao-You Fan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Han Ding
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao-Shuai Tang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Da-Yu Pan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jia-Xiao Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng-Yuan Zheng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong-Yu Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Heng Wu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ang Li
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, Henan Province, China
| | - Shi-Qing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Eve DJ, Sanberg PR, Buzanska L, Sarnowska A, Domanska-Janik K. Human Somatic Stem Cell Neural Differentiation Potential. Results Probl Cell Differ 2018; 66:21-87. [DOI: 10.1007/978-3-319-93485-3_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
5
|
Xue J, Yang J, O’Connor DM, Zhu C, Huo D, Boulis NM, Xia Y. Differentiation of Bone Marrow Stem Cells into Schwann Cells for the Promotion of Neurite Outgrowth on Electrospun Fibers. ACS APPLIED MATERIALS & INTERFACES 2017; 9:12299-12310. [PMID: 28322042 PMCID: PMC5489349 DOI: 10.1021/acsami.7b00882] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Seeding nerve guidance conduits with Schwann cells can improve the outcome of peripheral nerve injury repair. Bone marrow stem cells (BMSCs) represent a good choice of cell source as they can differentiate into Schwann cells under appropriate conditions. In this work, we systematically investigated the differentiation of BMSCs into Schwann cells on scaffolds comprising electrospun fibers. We changed the alignment, diameter, and surface properties of the fibers to optimize the differentiation efficiency. The uniaxial alignment of fibers not only promoted the differentiation of BMSCs into Schwann cells but also dictated the morphology and alignment of the derived cells. Coating the surface of aligned fibers with laminin further enhanced the differentiation and thus increased the secretion of neurotrophins. When co-cultured with PC12 cells or chick dorsal root ganglion, the as-derived Schwann cells were able to promote the outgrowth of neurites from cell bodies and direct their extension along the fibers, demonstrating the positive impacts of both the neurotrophic effect and the morphological contact guidance. This work offers a promising strategy for integrating fiber guidance with stem cell therapy to augment peripheral nerve injury repair.
Collapse
Affiliation(s)
- Jiajia Xue
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Junyu Yang
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Deirdre M. O’Connor
- Department of Neurosurgery, Emory University, Atlanta, Georgia 30322, United States
| | - Chunlei Zhu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Da Huo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Nicholas M. Boulis
- Department of Neurosurgery, Emory University, Atlanta, Georgia 30322, United States
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- School of Chemistry and Biochemistry, School of Chemical and Biological Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Corresponding Author:
| |
Collapse
|
6
|
Malignant transformation of bone marrow stromal cells induced by the brain glioma niche in rats. Mol Cell Biochem 2015; 412:1-10. [PMID: 26590986 DOI: 10.1007/s11010-015-2602-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 10/29/2015] [Indexed: 01/10/2023]
Abstract
Normal human embryonic stem cells (hESCs) can develop neoplastic cancer stem cell (CSC) properties after coculture with transformed hESCs in vitro. In the present study, the influence of the tumor microenvironment on malignant transformation of bone marrow stromal cells (BMSCs) was studied after allografting a mixture of enhanced green fluorescent protein (EGFP)-labeled BMSCs and C6 glioma cells into the rat brain to understand the influence of the cellular environment, especially the tumor environment, on the transformation of grafted BMSCs in the rat brain. We performed intracerebral transplantation in the rat brain using EGFP-labeled BMSCs coinjected with C6 tumor cells. After transplantation, the EGFP-labeled cells were isolated from the tumor using fluorescence-activated cell sorting, and the characteristics of the recovered cells were investigated. Glioma-specific biomarkers of the sorted cells and the biological characteristics of the tumors were analyzed. The BMSCs isolated from the cografts were transformed into glioma CSCs, as indicated by the marked expression of the glioma marker GFAP in glioma cells, and of Nestin and CD133 in neural stem cells and CSCs, as well as rapid cell growth, decreased level of the tumor suppressor gene p53, increased level of the oncogene murine double minute gene 2 (MDM2), and recapitulation of glioma tissues in the brain. These data suggest that BMSCs can be transformed into CSCs, which can be further directed toward glioma formation under certain conditions, supporting the notion that the tumor microenvironment is involved in transforming normal BMSCs into glial CSCs.
Collapse
|
7
|
Stem cell-based approaches to improve nerve regeneration: potential implications for reconstructive transplantation? Arch Immunol Ther Exp (Warsz) 2014; 63:15-30. [PMID: 25428664 DOI: 10.1007/s00005-014-0323-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/07/2014] [Indexed: 12/17/2022]
Abstract
Reconstructive transplantation has become a viable option to restore form and function after devastating tissue loss. Functional recovery is a key determinant of overall success and critically depends on the quality and pace of nerve regeneration. Several molecular and cell-based therapies have been postulated and tested in pre-clinical animal models to enhance nerve regeneration. Schwann cells remain the mainstay of research focus providing neurotrophic support and signaling cues for regenerating axons. Alternative cell sources such as mesenchymal stem cells and adipose-derived stromal cells have also been tested in pre-clinical animal models and in clinical trials due to their relative ease of harvest, rapid expansion in vitro, minimal immunogenicity, and capacity to integrate and survive within host tissues, thereby overcoming many of the challenges faced by culturing of human Schwann cells and nerve allografting. Induced pluripotent stem cell-derived Schwann cells are of particular interest since they can provide abundant, patient-specific autologous Schwann cells. The majority of experimental evidence on cell-based therapies, however, has been generated using stem cell-seeded nerve guides that were developed to enhance nerve regeneration across "gaps" in neural repair. Although primary end-to-end repair is the preferred method of neurorrhaphy in reconstructive transplantation, mechanistic studies elucidating the principles of cell-based therapies from nerve guidance conduits will form the foundation of further research employing stem cells in end-to-end repair of donor and recipient nerves. This review presents key components of nerve regeneration in reconstructive transplantation and highlights the pre-clinical studies that utilize stem cells to enhance nerve regeneration.
Collapse
|
8
|
Park YJ, Koh J, Gauna AE, Chen S, Cha S. Identification of regulatory factors for mesenchymal stem cell-derived salivary epithelial cells in a co-culture system. PLoS One 2014; 9:e112158. [PMID: 25402494 PMCID: PMC4234408 DOI: 10.1371/journal.pone.0112158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 10/13/2014] [Indexed: 01/12/2023] Open
Abstract
Patients with Sjögren’s syndrome or head and neck cancer patients who have undergone radiation therapy suffer from severe dry mouth (xerostomia) due to salivary exocrine cell death. Regeneration of the salivary glands requires a better understanding of regulatory mechanisms by which stem cells differentiate into exocrine cells. In our study, bone marrow-derived mesenchymal stem cells were co-cultured with primary salivary epithelial cells from C57BL/6 mice. Co-cultured bone marrow-derived mesenchymal stem cells clearly resembled salivary epithelial cells, as confirmed by strong expression of salivary gland epithelial cell-specific markers, such as alpha-amylase, muscarinic type 3 receptor, aquaporin-5, and cytokeratin 19. To identify regulatory factors involved in this differentiation, transdifferentiated mesenchymal stem cells were analyzed temporarily by two-dimensional-gel-electrophoresis, which detected 58 protein spots (>1.5 fold change, p<0.05) that were further categorized into 12 temporal expression patterns. Of those proteins only induced in differentiated mesenchymal stem cells, ankryin-repeat-domain-containing-protein 56, high-mobility-group-protein 20B, and transcription factor E2a were selected as putative regulatory factors for mesenchymal stem cell transdifferentiation based on putative roles in salivary gland development. Induction of these molecules was confirmed by RT-PCR and western blotting on separate sets of co-cultured mesenchymal stem cells. In conclusion, our study is the first to identify differentially expressed proteins that are implicated in mesenchymal stem cell differentiation into salivary gland epithelial cells. Further investigation to elucidate regulatory roles of these three transcription factors in mesenchymal stem cell reprogramming will provide a critical foundation for a novel cell-based regenerative therapy for patients with xerostomia.
Collapse
Affiliation(s)
- Yun-Jong Park
- Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Jin Koh
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, United States of America
| | - Adrienne E. Gauna
- Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Sixue Chen
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, United States of America
- Department of Biology, UF Genetics Institute, University of Florida, Gainesville, Florida, United States of America
- Genetics Institute, University of Florida, Gainesville, Florida, United States of America
| | - Seunghee Cha
- Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
9
|
Lo Furno D, Pellitteri R, Graziano ACE, Giuffrida R, Vancheri C, Gili E, Cardile V. Differentiation of human adipose stem cells into neural phenotype by neuroblastoma- or olfactory ensheathing cells-conditioned medium. J Cell Physiol 2013; 228:2109-2118. [PMID: 23589068 DOI: 10.1002/jcp.24386] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 04/05/2013] [Indexed: 01/12/2023]
Abstract
Olfactory ensheathing cells (OECs) are known to be capable of continuous neurogenesis throughout lifetime and are a source of multiple trophic factors important in central nervous system regeneration. B104 neuroblastoma cells are recognized to induce differentiation of neural stem cells into oligodendrocyte precursor cells. Therefore, the aim of this study was to verify if conditioned medium (CM) obtained from OECs or B104 cells was capable of inducing differentiation of adipose tissue-derived mesenchymal stem cells (AT-MSCs) to a neuronal phenotype. In order to this goal, immunocytochemical procedures and flow cytometry analysis were used and some neural markers, as nestin, protein gene product 9.5 (PGP 9.5), microtubule-associated protein 2 (MAP2), glial fibrillary acidic protein (GFAP), and neuron cell surface antigen (A2B5) were examined 24 h and 7 days after the treatment. The results showed that both OECs- or B104-CM treated AT-MSCs express markers of progenitor and mature neurons (nestin, PGP 9.5 and MAP2) in time-dependent manner, display morphological features resembling neuronal cells, and result negative for GFAP and A2B5, astrocyte and oligodendrocyte markers, respectively. This study demonstrated that AT-MSCs can be influenced by the environment, indicating that these cells can respond to environmental cues also versus a neuronal phenotype.
Collapse
Affiliation(s)
- Debora Lo Furno
- Department of Bio-medical Sciences, Section Physiology, University of Catania, Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
10
|
Li J, Qiao X, Yu M, Li F, Wang H, Guo W, Tian W. Secretory Factors From Rat Adipose Tissue Explants Promote Adipogenesis and Angiogenesis. Artif Organs 2013; 38:E33-45. [PMID: 24020992 DOI: 10.1111/aor.12162] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jie Li
- College of Life Science; Sichuan University; Chengdu China
- National Engineering Laboratory for Oral Regenerative Medicine; Sichuan University; Chengdu China
- State Key Laboratory of Oral Diseases; Sichuan University; Chengdu China
| | - Xiangchen Qiao
- National Engineering Laboratory for Oral Regenerative Medicine; Sichuan University; Chengdu China
- State Key Laboratory of Oral Diseases; Sichuan University; Chengdu China
| | - Mei Yu
- National Engineering Laboratory for Oral Regenerative Medicine; Sichuan University; Chengdu China
- State Key Laboratory of Oral Diseases; Sichuan University; Chengdu China
| | - Feng Li
- National Engineering Laboratory for Oral Regenerative Medicine; Sichuan University; Chengdu China
- State Key Laboratory of Oral Diseases; Sichuan University; Chengdu China
- Department of Oral and Maxillofacial Surgery; West China School of Stomatology; Sichuan University; Chengdu China
| | - Hang Wang
- National Engineering Laboratory for Oral Regenerative Medicine; Sichuan University; Chengdu China
- State Key Laboratory of Oral Diseases; Sichuan University; Chengdu China
- Department of Oral and Maxillofacial Surgery; West China School of Stomatology; Sichuan University; Chengdu China
| | - Weihua Guo
- National Engineering Laboratory for Oral Regenerative Medicine; Sichuan University; Chengdu China
- State Key Laboratory of Oral Diseases; Sichuan University; Chengdu China
- Department of Pedodontics; West China School of Stomatology; Sichuan University; Chengdu China
| | - Weidong Tian
- National Engineering Laboratory for Oral Regenerative Medicine; Sichuan University; Chengdu China
- State Key Laboratory of Oral Diseases; Sichuan University; Chengdu China
- Department of Oral and Maxillofacial Surgery; West China School of Stomatology; Sichuan University; Chengdu China
| |
Collapse
|
11
|
Expression of the chitinase family glycoprotein YKL-40 in undifferentiated, differentiated and trans-differentiated mesenchymal stem cells. PLoS One 2013; 8:e62491. [PMID: 23671604 PMCID: PMC3650021 DOI: 10.1371/journal.pone.0062491] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 03/22/2013] [Indexed: 12/19/2022] Open
Abstract
The glycoprotein YKL-40 (CHI3L1) is a secreted chitinase family protein that induces angiogenesis, cell survival, and cell proliferation, and plays roles in tissue remodeling and immune regulation. It is expressed primarily in cells of mesenchymal origin, is overexpressed in numerous aggressive carcinomas and sarcomas, but is rarely expressed in normal ectodermal tissues. Bone marrow-derived mesenchymal stem cells (MSCs) can be induced to differentiate into various mesenchymal tissues and trans-differentiate into some non-mesenchymal cell types. Since YKL-40 has been used as a mesenchymal marker, we followed YKL-40 expression as undifferentiated MSCs were induced to differentiate into bone, cartilage, and neural phenotypes. Undifferentiated MSCs contain significant levels of YKL-40 mRNA but do not synthesize detectable levels of YKL-40 protein. MSCs induced to differentiate into chondrocytes and osteocytes soon began to express and secrete YKL-40 protein, as do ex vivo cultured chondrocytes and primary osteocytes. In contrast, MSCs induced to trans-differentiate into neurons did not synthesize YKL-40 protein, consistent with the general absence of YKL-40 protein in normal CNS parenchyma. However, these trans-differentiated neurons retained significant levels of YKL-40 mRNA, suggesting the mechanisms which prevented YKL-40 translation in undifferentiated MSCs remained in place, and that these trans-differentiated neurons differ in at least this way from neurons derived from neuronal stem cells. Utilization of a differentiation protocol containing β-mercaptoethanol resulted in cells that expressed significant amounts of intracellular YKL-40 protein that was not secreted, which is not seen in normal cells. Thus the synthesis of YKL-40 protein is a marker for MSC differentiation into mature mesenchymal phenotypes, and the presence of untranslated YKL-40 mRNA in non-mesenchymal cells derived from MSCs reflects differences between differentiated and trans-differentiated phenotypes.
Collapse
|
12
|
Rutten MJ, Janes MA, Chang IR, Gregory CR, Gregory KW. Development of a functional schwann cell phenotype from autologous porcine bone marrow mononuclear cells for nerve repair. Stem Cells Int 2012; 2012:738484. [PMID: 22792117 PMCID: PMC3388598 DOI: 10.1155/2012/738484] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 03/29/2012] [Indexed: 01/10/2023] Open
Abstract
Adult bone marrow mononuclear cells (BM-MNCs) are a potential resource for making Schwann cells to repair damaged peripheral nerves. However, many methods of producing Schwann-like cells can be laborious with the cells lacking a functional phenotype. The objective of this study was to develop a simple and rapid method using autologous BM-MNCs to produce a phenotypic and functional Schwann-like cell. Adult porcine bone marrow was collected and enriched for BM-MNCs using a SEPAX device, then cells cultured in Neurobasal media, 4 mM L-glutamine and 20% serum. After 6-8 days, the cultures expressed Schwann cell markers, S-100, O4, GFAP, were FluoroMyelin positive, but had low p75(NGF) expression. Addition of neuregulin (1-25 nM) increased p75(NGF) levels at 24-48 hrs. We found ATP dose-dependently increased intracellular calcium [Ca(2+)](i), with nucleotide potency being UTP = ATP > ADP > AMP > adenosine. Suramin blocked the ATP-induced [Ca(2+)](i) but α, β,-methylene-ATP had little effect suggesting an ATP purinergic P2Y2 G-protein-coupled receptor is present. Both the Schwann cell markers and ATP-induced [Ca(2+)](i) sensitivity decreased in cells passaged >20 times. Our studies indicate that autologous BM-MNCs can be induced to form a phenotypic and functional Schwann-like cell which could be used for peripheral nerve repair.
Collapse
Affiliation(s)
- Michael J. Rutten
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | - Michael Ann Janes
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
| | - Ivy R. Chang
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
| | - Cynthia R. Gregory
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
- Oregon Biomedical Engineering Institute, 25999 SW Canyon Creek Rd., Wilsonville, OR 97070, USA
- Portland VA Medical Center, 3710 SW U.S. Veterans Hospital Rd., Portland, OR 97239, USA
| | - Kenton W. Gregory
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
- Oregon Biomedical Engineering Institute, 25999 SW Canyon Creek Rd., Wilsonville, OR 97070, USA
| |
Collapse
|
13
|
Tomita K, Madura T, Mantovani C, Terenghi G. Differentiated adipose-derived stem cells promote myelination and enhance functional recovery in a rat model of chronic denervation. J Neurosci Res 2012; 90:1392-402. [PMID: 22419645 DOI: 10.1002/jnr.23002] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 11/07/2011] [Indexed: 12/13/2022]
Abstract
Transplantation of autologous Schwann cells (SCs) is a promising approach for treating various peripheral nerve disorders, including chronic denervation. However, given their drawbacks, such as invasive biopsy and lengthy culture in vitro, alternative cell sources would be needed. Adipose-derived stem cells (ASCs) are a candidate, and in this study rat ASCs transdifferentiated into a SC phenotype (dASC) cocultured with dorsal root ganglion neurons were shown to associate with neurites and to express myelin basic protein (MBP)-positive myelin protein. Furthermore, dASCs transplanted into a chronically denervated rat common peroneal nerve survived for at least for 10 weeks, maintaining their differentiated state. Immunohistochemical analysis revealed that transplanted dASCs associated with regenerating axons, forming MBP-/protein zero-positive myelin sheaths. The cell survival and myelin expression assessed by double labelling with S100 and glial fibrillary acidic protein were similar between the dASC- and SC-transplanted nerves. Importantly, transplantation of dASCs resulted in dramatically improved motor functional recovery and nerve regeneration, with a level comparable to, or even superior to, transplantation of SCs. In conclusion, dASCs are capable of myelinating axons in vivo and enhancing functional outcome after chronic denervation.
Collapse
Affiliation(s)
- Koichi Tomita
- Blond McIndoe Laboratories, Regenerative Biomedicine, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| | | | | | | |
Collapse
|
14
|
Bonilla C, Zurita M, Otero L, Aguayo C, Rico MA, Rodríguez A, Vaquero J. Failure of Delayed Intravenous Administration of Bone Marrow Stromal Cells after Traumatic Brain Injury. J Neurotrauma 2012; 29:394-400. [DOI: 10.1089/neu.2011.2101] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Celia Bonilla
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Mercedes Zurita
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Laura Otero
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Concepción Aguayo
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Miguel A. Rico
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Alicia Rodríguez
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Jesús Vaquero
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
- Service of Neurosurgery, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| |
Collapse
|
15
|
Kwon EB, Lee JY, Piao S, Kim IG, Ra JC, Lee JY. Comparison of human muscle-derived stem cells and human adipose-derived stem cells in neurogenic trans-differentiation. Korean J Urol 2011; 52:852-7. [PMID: 22216399 PMCID: PMC3246519 DOI: 10.4111/kju.2011.52.12.852] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 10/10/2011] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Erectile dysfunction (ED) remains a major complication from cavernous nerve injury during radical prostatectomy. Recently, stem cell treatment for ED has been widely reported. This study was conducted to investigate the availability, differentiation into functional cells, and potential of human muscle-derived stem cells (hMDSCs) and human adipose-derived stem cells (hADSCs) for ED treatment. MATERIALS AND METHODS We compared the neural differentiation of hMDSCs and hADSCs. Human muscle and adipose tissues were digested with collagenase, followed by filtering and centrifugation. For neural induction, isolated hMDSCs and hADSCs were incubated in neurobasal media containing forskolin, laminin, basic-fibroblast growth factor, and epidermal growth factor for 5 days. Following neural induction, hMDSCs and hADSCs were differentiated into neural cells, including neurons and glia, in vitro. RESULTS In neural differentiated hMDSCs (d-hMDSCs) and differentiated hADSCs (d-hADSCs), neural stem cell marker (nestin) showed a significant decrease by immunocytochemistry, and neuronal marker (β-tubulin III) and glial marker (GFAP) showed a significant increase, compared with primary hMDSCs and hADSCs. Real-time chain reaction analysis and Western blotting demonstrated significantly elevated levels of mRNA and protein of β-tubulin III and GFAP in d-hADSCs compared with d-hMDSCs. CONCLUSIONS We demonstrated that hMDSCs and hADSCs can be induced to undergo phenotypic and molecular changes consistent with neurons. The neural differentiation capacity of hADSCs was better than that of hMDSCs.
Collapse
Affiliation(s)
- Eun Bi Kwon
- Department of Urology, College of Medicine, The Catholic University of Korea, Ltd. Seoul, Korea
| | | | | | | | | | | |
Collapse
|
16
|
Hong SQ, Zhang HT, You J, Zhang MY, Cai YQ, Jiang XD, Xu RX. Comparison of transdifferentiated and untransdifferentiated human umbilical mesenchymal stem cells in rats after traumatic brain injury. Neurochem Res 2011; 36:2391-400. [PMID: 21877237 DOI: 10.1007/s11064-011-0567-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 07/26/2011] [Accepted: 07/27/2011] [Indexed: 12/23/2022]
Abstract
Transdifferentiated and untransdifferentiated mesenchymal stem cells (MSCs) have shown therapeutic benefits in central nervous system (CNS) injury. However, it is unclear which would be more appropriate for transplantation. To address this question, we transplanted untransdifferentiated human umbilical mesenchymal stem cells (HUMSCs) and transdifferentiated HUMSCs (HUMSC-derived neurospheres, HUMSC-NSs) into a rat model of traumatic brain injury. Cognitive function, cell survival and differentiation, brain tissue morphology and neurotrophin expression were compared between groups. Significant improvements in cognitive function and brain tissue morphology were seen in the HUMSCs group compared with HUMSC-NSs group, which was accompanied by increased neurotrophin expression. Moreover, only few grafted cells survived in both the HUMSCs and HUMSC-NSs groups, with very few of the cells differentiating into neural-like cells. These findings indicate that HUMSCs are more appropriate for transplantation and their therapeutic benefits may be associated with neuroprotection rather than cell replacement.
Collapse
Affiliation(s)
- Sun-Quan Hong
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Zurita M, Otero L, Aguayo C, Bonilla C, Ferreira E, Parajón A, Vaquero J. Cell therapy for spinal cord repair: optimization of biologic scaffolds for survival and neural differentiation of human bone marrow stromal cells. Cytotherapy 2011; 12:522-37. [PMID: 20465485 DOI: 10.3109/14653241003615164] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND AIMS The suppression of cell apoptosis using a biodegradable scaffold to replace the missing or altered extracellular matrix (ECM) could increase the survival of transplanted cells and thus increase the effectiveness of cell therapy. METHODS We studied the best conditions for the proliferation and differentiation of human bone marrow stromal cells (hBMSC) when cultured on different biologic scaffolds derived from fibrin and blood plasma, and analyzed the best concentrations of fibrinogen, thrombin and calcium chloride for favoring cell survival. The induction of neural differentiation of hBMSC was done by adding to these scaffolds different growth factors, such as nerve growth factor (NGF), brain-derived-neurotrophic factor (BDNF) and retinoic acid (RA), at concentrations of 100 ng/mL (NGF and BDNF) and 1 micro/mL (RA), over 7 days. RESULTS Although both types of scaffold allowed survival and neural differentiation of hBMSC, the results showed a clear superiority of platelet-rich plasma (PRP) scaffolds, mainly after BDNF administration, allowing most of the hBMSC to survive and differentiate into a neural phenotype. CONCLUSIONS Given that clinical trials for spinal cord injury using hBMSC are starting, these findings may have important clinical applications.
Collapse
Affiliation(s)
- Mercedes Zurita
- Neuroscience Research Unit of Surgical Research Service, Hospital Puerta de Hierro-Majadahonda, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
18
|
Maria OM, Tran SD. Human mesenchymal stem cells cultured with salivary gland biopsies adopt an epithelial phenotype. Stem Cells Dev 2011; 20:959-67. [PMID: 21187001 DOI: 10.1089/scd.2010.0214] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Sjogren's syndrome and radiotherapy for head and neck cancer result in severe xerostomia and irreversible salivary gland damage for which no effective treatment is currently available. Cell culture methods of primary human salivary gland epithelial cells (huSGs) are slow and cannot provide a sufficient number of cells. In addition, the majority of cultured huSGs are of a ductal phenotype and thus not fluid/saliva secretory cells. Some reports indicated that mesenchymal stem cells (MSCs) possessed the potential to differentiate into epithelial cells. To test this hypothesis with huSGs, a coculture system containing 2 chambers separated by a polyester membrane was used to study the capacity of human MSCs to adopt an epithelial phenotype when cocultured with human salivary gland biopsies. Results were that 20%-40% of cocultured MSCs expressed tight junction proteins [claudin-1 (CLDN-1), -2, -3, and -4; occludin; junctional adhesion molecule-A; and zonula occludens-1] as well as other epithelial markers [aquaporin-5, α-amylase (α-AMY), and E-cadherin], and generated a higher transepithelial electrical resistance. Electron microscopy demonstrated that these MSCs had comparable cellular structures to huSGs, such as tight junction structures and numerous secretory granules. Quantitative real time (RT)-polymerase chain reaction revealed an upregulation of several salivary genes (aquaporin-5, AMY, and CLDN-2). Moreover, the amounts of α-AMY detected in cocultured MSCs were comparable to those detected in huSGs control cultures. These data suggest that cocultured MSCs can demonstrate a temporary change into a salivary gland acinar phenotype.
Collapse
Affiliation(s)
- Ola M Maria
- Faculty of Dentistry, McGill University, Montreal, Canada
| | | |
Collapse
|
19
|
Zhang HT, Luo J, Sui LS, Ma X, Yan ZJ, Lin JH, Wang YS, Chen YZ, Jiang XD, Xu RX. Effects of differentiated versus undifferentiated adipose tissue-derived stromal cell grafts on functional recovery after spinal cord contusion. Cell Mol Neurobiol 2009; 29:1283-92. [PMID: 19533335 PMCID: PMC11505793 DOI: 10.1007/s10571-009-9424-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Accepted: 06/03/2009] [Indexed: 02/06/2023]
Abstract
Controversies exist concerning the need for mesenchymal stromal cells (MSCs) to be transdifferentiated prior to their transplantation. In the present study, we compared the results of grafting into the rat contused spinal cord undifferentiated, adipose tissue-derived stromal cells (uADSCs) versus ADSCs induced by two different protocols to form differentiated nervous tissue. Using Basso, Beattie, and Bresnahan scores and grid tests, we found that three cell-treated groups, including uADSCs-treated, dADSCs induced by Protocol 1 (dADSC-P1)-treated, and dADSCs induced by Protocol 2 (dADSC-P2)-treated groups, significantly improved locomotor functional recovery in SCI rats, compared with the saline-treated group. Furthermore, functional recovery was better in the uADSC-treated and dADSC-P2-treated groups than in the dADSC-P1-treated group at week 12 postinjury (P < 0.05 for dADSC-P1 group vs. uADSCs or dADSC-P2 groups). Although both protocols could induce high percentages of cells expressing neural markers in vitro, few BrdU-labeled cells survived at the injury sites in the three cell-treated groups, and only a small percentage of BrdU-positive cells expressed neural markers. On the other hand, the number of NF200-positive axons in the uADSC-treated and dADSC-P2-treated groups was significantly larger than those in the dADSC-P1-treated and saline-treated control groups. Our results indicate that ADSCs are able to differentiate into neural-like cells in vitro and in vivo. However, neural differentiated ADSCs did not result in better functional recovery than undifferentiated ones, following SCI. In vitro neural transdifferentiation of ADSCs might therefore not be a necessary pretransplantation step. Furthermore, cellular replacement or integration might not contribute to the functional recovery of the injured spinal cord.
Collapse
Affiliation(s)
- Hong-Tian Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Jie Luo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Li-Sen Sui
- Department of Neurosurgery, Guangdong Hospital of Traditional Chinese Medicine, 510120 Guangzhou, China
| | - Xu Ma
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Zhong-Jie Yan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Jian-Hao Lin
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Yu-Sheng Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Yi-Zhao Chen
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Xiao-Dan Jiang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Ru-Xiang Xu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
- Department of Neurosurgery, The Military General Hospital of Beijing PLA, 100700 Beijing, China
| |
Collapse
|
20
|
Zhang N, Wimmer J, Qian SJ, Chen WS. Stem Cells: Current Approach and Future Prospects in Spinal Cord Injury Repair. Anat Rec (Hoboken) 2009; 293:519-30. [DOI: 10.1002/ar.21025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Zhang YQ, Zeng X, He LM, Ding Y, Li Y, Zeng YS. NT-3 gene modified Schwann cells promote TrkC gene modified mesenchymal stem cells to differentiate into neuron-like cells in vitro. Anat Sci Int 2009; 85:61-7. [DOI: 10.1007/s12565-009-0056-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 07/03/2009] [Indexed: 01/01/2023]
|
22
|
Characterisation of human mesenchymal stem cells following differentiation into Schwann cell-like cells. Neurosci Res 2009; 64:41-9. [PMID: 19428682 DOI: 10.1016/j.neures.2009.01.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 01/07/2009] [Accepted: 01/16/2009] [Indexed: 11/22/2022]
Abstract
Cell-based therapies provide a clinically applicable and available alternative to nerve autografts. Our previous studies have characterised rat-derived mesenchymal stem cells (MSC) and here we have investigated the phenotypic, molecular and functional characteristics of human-derived MSC (hMSC) differentiated along a Schwann cell lineage. The hMSC were isolated from healthy human donors and the identity of the undifferentiated hMSC was confirmed by the detection of MSC specific cells surface markers. The hMSC were differentiated along a glial cell lineage using an established cocktail of growth factors including glial growth factor-2. Following differentiation, the hMSC expressed the key Schwann cell (SC) markers at both the transcriptional and translational level. More importantly, we show the functional effect of hMSC on neurite outgrowth using an in vitro co-culture model system with rat-derived primary sensory neurons. The number of DRG sprouting neurites was significantly enhanced in the presence of differentiated hMSC; neurite length and density (branching) were also increased. These results provide evidence that hMSC can undergo molecular, morphological and functional changes to adopt a SC-like behaviour and, therefore, could be suitable as SC substitutes for nerve repair in clinical applications.
Collapse
|
23
|
Marcoli M, Candiani S, Tonachini L, Monticone M, Mastrogiacomo M, Ottonello A, Cervetto C, Paluzzi P, Maura G, Pestarino M, Cancedda R, Castagnola P. In vitro modulation of gamma amino butyric acid (GABA) receptor expression by bone marrow stromal cells. Pharmacol Res 2008; 57:374-82. [PMID: 18467116 DOI: 10.1016/j.phrs.2008.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 03/26/2008] [Accepted: 03/27/2008] [Indexed: 01/14/2023]
Abstract
Bone marrow stromal cells (BMSC) have the capability to undergo a change of morphology, reminiscent of neuronal cells, after exposure to an inductive medium. These induced BMSC-derived neuron-like (BDNL) cells express several neuronal markers, including Microtubule-Associated Protein Tau, Neurofilament M, and Nestin as revealed by immunocytochemistry analysis. To assess whether the induction process has possible functional relevance, we have focused our attention on the expression of neurotransmitter receptors. In particular, we show that the expression of GABA(A) subunits alpha1, beta2/3, epsilon and GABA(B1) mRNAs is greatly enhanced in BMSC by the induction treatment. Similar results were obtained from rat skin fibroblasts subjected to the same induction protocol, with the exception for the GABA(B2) transcript that was expressed only by BMSC and BDNL. The presence of both GABA(B1) and GABA(B2) subunits in BDNL cells suggests that functional GABA(B) receptors might be assembled: we indeed found that a functional GABA(B) receptor, negatively linked to cyclic AMP production, is expressed in BDNL. Therefore, we suggest that BMSC can be converted into cells equipped with appropriate receptors coupled to transduction mechanisms, potentially responding to a specific neurotransmitter.
Collapse
Affiliation(s)
- Manuela Marcoli
- Dip. Medicina Sperimentale, Sezione di Farmacologia e Tossicologia, Università degli Studi di, Genova, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|