1
|
Li Z, Kegui H, Piao W, Xuejiu W, Lim KT, Jin H. PAI-1 transfected-conditioned media promotes osteogenic differentiation of hBMSCs. Cell Biol Int 2024. [PMID: 38654436 DOI: 10.1002/cbin.12166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 02/28/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Reconstruction of injured bone remains challenging in the clinic owing to the lack of suitable bone grafts. The utilization of PAI-1 transfected-conditioned media (P-CM) has demonstrated its ability to facilitate the differentiation process of mesenchymal stem cells (MSCs), potentially serving as a crucial mediator in tissue regeneration. This research endeavored to explore the therapeutic potential of P-CM concerning the differentiation of human bone marrow mesenchymal stem cells (hBMSCs). To assess new bone formation, a rat calvaria critical defect model was employed, while in vitro experiments involved the use of the alizarin Red-S mineral induction test. In the rat calvaria critical defect model, P-CM treatment resulted in significan new bone formation. In vitro, P-CM treated hBMSCs displayed robust osteogenesis compared to the control group, as demonstrated by the mineral induction test using alizarin Red-S. P-CM with hydroxyapatite/β-tricalcium phosphate/fibrin gel treatment significantly exhibited new bone formation, and the expression of osteogenic associated markers was enhanced in the P-CM-treated group. In conclusion, results demonstrate that P-CM treatment significantly enhanced the osteogenic differantiation efficiency and new bone formation, thus could be used as an ideal therapeutic biomolecule for constructing bone-specific implants, especially for orthopedic and dental applications.
Collapse
Affiliation(s)
- Zhang Li
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Hou Kegui
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wang Piao
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wang Xuejiu
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, South Korea
| | - Hexiu Jin
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Karacan I, Milthorpe B, Ben-Nissan B, Santos J. Stem Cells and Proteomics in Biomaterials and Biomedical Applications. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2022:125-157. [DOI: 10.1007/978-981-16-7435-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Okolicsanyi RK, Oikari LE, Yu C, Griffiths LR, Haupt LM. Heparan Sulfate Proteoglycans as Drivers of Neural Progenitors Derived From Human Mesenchymal Stem Cells. Front Mol Neurosci 2018; 11:134. [PMID: 29740281 PMCID: PMC5928449 DOI: 10.3389/fnmol.2018.00134] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/03/2018] [Indexed: 01/19/2023] Open
Abstract
Background: Due to their relative ease of isolation and their high ex vivo and in vitro expansive potential, human mesenchymal stem cells (hMSCs) are an attractive candidate for therapeutic applications in the treatment of brain injury and neurological diseases. Heparan sulfate proteoglycans (HSPGs) are a family of ubiquitous proteins involved in a number of vital cellular processes including proliferation and stem cell lineage differentiation. Methods: Following the determination that hMSCs maintain neural potential throughout extended in vitro expansion, we examined the role of HSPGs in mediating the neural potential of hMSCs. hMSCs cultured in basal conditions (undifferentiated monolayer cultures) were found to co-express neural markers and HSPGs throughout expansion with modulation of the in vitro niche through the addition of exogenous HS influencing cellular HSPG and neural marker expression. Results: Conversion of hMSCs into hMSC Induced Neurospheres (hMSC IN) identified distinctly localized HSPG staining within the spheres along with altered gene expression of HSPG core protein and biosynthetic enzymes when compared to undifferentiated hMSCs. Conclusion: Comparison of markers of pluripotency, neural self-renewal and neural lineage specification between hMSC IN, hMSC and human neural stem cell (hNSC H9) cultures suggest that in vitro generated hMSC IN may represent an intermediary neurogenic cell type, similar to a common neural progenitor cell. In addition, this data demonstrates HSPGs and their biosynthesis machinery, are associated with hMSC IN formation. The identification of specific HSPGs driving hMSC lineage-specification will likely provide new markers to allow better use of hMSCs in therapeutic applications and improve our understanding of human neurogenesis.
Collapse
Affiliation(s)
- Rachel K Okolicsanyi
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lotta E Oikari
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Chieh Yu
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Larisa M Haupt
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
4
|
Zeng X, Ma YH, Chen YF, Qiu XC, Wu JL, Ling EA, Zeng YS. Autocrine fibronectin from differentiating mesenchymal stem cells induces the neurite elongation in vitro and promotes nerve fiber regeneration in transected spinal cord injury. J Biomed Mater Res A 2016; 104:1902-11. [PMID: 26991461 PMCID: PMC5101622 DOI: 10.1002/jbm.a.35720] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 03/02/2016] [Accepted: 03/11/2016] [Indexed: 01/08/2023]
Abstract
Extracellular matrix (ECM) expression is temporally and spatially regulated during the development of stem cells. We reported previously that fibronectin (FN) secreted by bone marrow mesenchymal stem cells (MSCs) was deposited on the surface of gelatin sponge (GS) soon after culture. In this study, we aimed to assess the function of accumulated FN on neuronal differentiating MSCs as induced by Schwann cells (SCs) in three dimensional transwell co‐culture system. The expression pattern and amount of FN of differentiating MSCs was examined by immunofluorescence, Western blot and immunoelectron microscopy. The results showed that FN accumulated inside GS scaffold, although its mRNA expression in MSCs was progressively decreased during neural induction. MSC‐derived neuron‐like cells showed spindle‐shaped cell body and long extending processes on FN‐decorated scaffold surface. However, after blocking of FN function by application of monoclonal antibodies, neuron‐like cells showed flattened cell body with short and thick neurites, together with decreased expression of integrin β1. In vivo transplantation study revealed that autocrine FN significantly facilitated endogenous nerve fiber regeneration in spinal cord transection model. Taken together, the present results showed that FN secreted by MSCs in the early stage accumulated on the GS scaffold and promoted the neurite elongation of neuronal differentiating MSCs as well as nerve fiber regeneration after spinal cord injury. This suggests that autocrine FN has a dynamic influence on MSCs in a three dimensional culture system and its potential application for treatment of traumatic spinal cord injury. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1902–1911, 2016.
Collapse
Affiliation(s)
- Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
- Department of Histology and Embryology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yuan-Feng Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xue-Cheng Qiu
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
| | - Jin-Lang Wu
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
- Co-innovation Center of Neuroregeneration, Jiangsu, 226019, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
5
|
Integration of donor mesenchymal stem cell-derived neuron-like cells into host neural network after rat spinal cord transection. Biomaterials 2015; 53:184-201. [DOI: 10.1016/j.biomaterials.2015.02.073] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/08/2015] [Accepted: 02/15/2015] [Indexed: 12/27/2022]
|
6
|
Zhang RP, Xu C, Liu Y, Li JD, Xie J. Visual bone marrow mesenchymal stem cell transplantation in the repair of spinal cord injury. Neural Regen Res 2015; 10:404-11. [PMID: 25878588 PMCID: PMC4396102 DOI: 10.4103/1673-5374.153688] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2015] [Indexed: 12/23/2022] Open
Abstract
An important factor in improving functional recovery from spinal cord injury using stem cells is maximizing the number of transplanted cells at the lesion site. Here, we established a contusion model of spinal cord injury by dropping a weight onto the spinal cord at T7-8. Superparamagnetic iron oxide-labeled bone marrow mesenchymal stem cells were transplanted into the injured spinal cord via the subarachnoid space. An outer magnetic field was used to successfully guide the labeled cells to the lesion site. Prussian blue staining showed that more bone marrow mesenchymal stem cells reached the lesion site in these rats than in those without magnetic guidance or superparamagnetic iron oxide labeling, and immunofluorescence revealed a greater number of complete axons at the lesion site. Moreover, the Basso, Beattie and Bresnahan (BBB) locomotor rating scale scores were the highest in rats with superparamagnetic labeling and magnetic guidance. Our data confirm that superparamagnetic iron oxide nanoparticles effectively label bone marrow mesenchymal stem cells and impart sufficient magnetism to respond to the external magnetic field guides. More importantly, superparamagnetic iron oxide-labeled bone marrow mesenchymal stem cells can be dynamically and non-invasively tracked in vivo using magnetic resonance imaging. Superparamagnetic iron oxide labeling of bone marrow mesenchymal stem cells coupled with magnetic guidance offers a promising avenue for the clinical treatment of spinal cord injury.
Collapse
Affiliation(s)
- Rui-ping Zhang
- Department of Radiology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Cheng Xu
- Department of Radiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi Province, China
| | - Yin Liu
- Department of Radiology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jian-ding Li
- Department of Radiology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jun Xie
- Department of Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
7
|
Du J, Gao X, Deng L, Chang N, Xiong H, Zheng Y. Transfection of the glial cell line-derived neurotrophic factor gene promotes neuronal differentiation. Neural Regen Res 2014; 9:33-40. [PMID: 25206741 PMCID: PMC4146314 DOI: 10.4103/1673-5374.125327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2013] [Indexed: 02/01/2023] Open
Abstract
Glial cell line-derived neurotrophic factor recombinant adenovirus vector-transfected bone marrow mesenchymal stem cells were induced to differentiate into neuron-like cells using inductive medium containing retinoic acid and epidermal growth factor. Cell viability, microtubule-associated protein 2-positive cell ratio, and the expression levels of glial cell line-derived neurotrophic factor, nerve growth factor and growth-associated protein-43 protein in the supernatant were significantly higher in glial cell line-derived neurotrophic factor/bone marrow mesenchymal stem cells compared with empty virus plasmid-transfected bone marrow mesenchymal stem cells. Furthermore, microtubule-associated protein 2, glial cell line-derived neurotrophic factor, nerve growth factor and growth-associated protein-43 mRNA levels in cell pellets were statistically higher in glial cell line-derived neurotrophic factor/bone marrow mesenchymal stem cells compared with empty virus plasmid-transfected bone marrow mesenchymal stem cells. These results suggest that glial cell line-derived neurotrophic factor/bone marrow mesenchymal stem cells have a higher rate of induction into neuron-like cells, and this enhanced differentiation into neuron-like cells may be associated with up-regulated expression of glial cell line-derived neurotrophic factor, nerve growth factor and growth-associated protein-43.
Collapse
Affiliation(s)
- Jie Du
- Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China ; Department of Anatomy, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Xiaoqing Gao
- Research Center for Preclinical Medicine, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Li Deng
- Research Center for Preclinical Medicine, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Nengbin Chang
- Department of Anatomy, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Huailin Xiong
- Department of Anatomy, Luzhou Medical College, Luzhou, Sichuan Province, China
| | - Yu Zheng
- Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
8
|
Wang Y, Peng W, Liu X, Zhu M, Sun T, Peng Q, Zeng Y, Feng B, Zhi W, Weng J, Wang J. Study of bilineage differentiation of human-bone-marrow-derived mesenchymal stem cells in oxidized sodium alginate/N-succinyl chitosan hydrogels and synergistic effects of RGD modification and low-intensity pulsed ultrasound. Acta Biomater 2014; 10:2518-2528. [PMID: 24394634 DOI: 10.1016/j.actbio.2013.12.052] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 12/05/2013] [Accepted: 12/26/2013] [Indexed: 12/15/2022]
Abstract
The level of formation of new bone and vascularization in bone tissue engineering scaffold implants is considered as a critical factor for clinical application. In this study, an approach using an RGD-grafted oxidized sodium alginate/N-succinyl chitosan (RGD-OSA/NSC) hydrogel as a scaffold and low-intensity pulsed ultrasound (LIPUS) as mechanical stimulation was proposed to achieve a high level of formation of new bone and vascularization. An in vitro study of endothelial and osteogenic differentiations of human-bone-marrow-derived mesenchymal stem cells (hMSCs) was conducted to evaluate it. The results showed that RGD-OSA/NSC composite hydrogels presented good biological properties in attachment, proliferation and differentiation of cells. The MTT cell viability assay showed that the total number of cells increased more significantly in the LIPUS-stimulated groups with RGD than that in the control ones; similar results were obtained for alkaline phosphatase activity/staining and mineralized nodule formation assay of osteogenic induction and immunohistochemical test of endothelial induction. The positive synergistic effect of LIPUS and RGD on the enhancement of proliferation and differentiation of hMSCs was observed. These findings suggest that the hybrid use of RGD modification and LIPUS might provide one approach to achieve a high level of formation of new bone and vascularization in bone tissue engineering scaffold implants.
Collapse
Affiliation(s)
- Yingying Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Wenzhen Peng
- Department of Biochemistry and Molecular Biology, College of Basic and Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xia Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Minghua Zhu
- Sichuan Centre for Disease Control and Prevention, Chengdu 610041, People's Republic of China
| | - Tao Sun
- Sichuan Centre for Disease Control and Prevention, Chengdu 610041, People's Republic of China
| | - Qiang Peng
- Sichuan Centre for Disease Control and Prevention, Chengdu 610041, People's Republic of China
| | - Yi Zeng
- Sichuan Centre for Disease Control and Prevention, Chengdu 610041, People's Republic of China
| | - Bo Feng
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Wei Zhi
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Jie Weng
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Jianxin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China.
| |
Collapse
|
9
|
Nakano R, Edamura K, Sugiya H, Narita T, Okabayashi K, Moritomo T, Teshima K, Asano K, Nakayama T. Evaluation of mRNA expression levels and electrophysiological function of neuron-like cells derived from canine bone marrow stromal cells. Am J Vet Res 2014; 74:1311-20. [PMID: 24066915 DOI: 10.2460/ajvr.74.10.1311] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To investigate the in vitro differentiation of canine bone marrow stromal cells (BMSCs) into functional, mature neurons. SAMPLE Bone marrow from 6 adult dogs. PROCEDURES BMSCs were isolated from bone marrow and chemically induced to develop into neurons. The morphology of the BMSCs during neuronal induction was monitored, and immunocytochemical analyses for neuron markers were performed after the induction. Real-time PCR methods were used to evaluate the mRNA expression levels of markers for neural stem or progenitor cells, neurons, and ion channels, and western blotting was used to assess the expression of neuronal proteins before and after neuronal induction. The electrophysiological properties of the neuron-like cells induced from canine BMSCs were evaluated with fluorescent dye to monitor Ca(2)+ influx. RESULTS Canine BMSCs developed a neuron-like morphology after neuronal induction. Immunocytochemical analysis revealed that these neuron-like cells were positive for neuron markers. After induction, the cells' mRNA expression levels of almost all neuron and ion channel markers increased, and the protein expression levels of nestin and neurofilament-L increased significantly. However, the neuron-like cells derived from canine BMSCs did not have the Ca(2)+ influx characteristic of spiking neurons. CONCLUSIONS AND CLINICAL RELEVANCE Although canine BMSCs had neuron-like morphological and biochemical properties after induction, they did not develop the electrophysiological characteristics of neurons. Thus, these results have suggested that canine BMSCs could have the capacity to differentiate into a neuronal lineage, but the differentiation protocol used may have been insufficient to induce development into functional neurons.
Collapse
Affiliation(s)
- Rei Nakano
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Jeon KJ, Park SH, Shin JW, Kang YG, Hyun JS, Oh MJ, Kim SY, Shin JW. Combined effects of flow-induced shear stress and micropatterned surface morphology on neuronal differentiation of human mesenchymal stem cells. J Biosci Bioeng 2014; 117:242-247. [DOI: 10.1016/j.jbiosc.2013.08.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 07/31/2013] [Accepted: 08/02/2013] [Indexed: 01/13/2023]
|
11
|
Wang L, Lu M. Regulation and direction of umbilical cord blood mesenchymal stem cells to adopt neuronal fate. Int J Neurosci 2013; 124:149-59. [PMID: 23879374 DOI: 10.3109/00207454.2013.828055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Umbilical cord blood mesenchymal stem cells (UCB-MSCs) transplantation is becoming a promising and attractive cell-based treatment modality for repairing the damaged central nervous system due to its advantages of low immunogenicity, wide range of sources, and less ethical controversy. One of the limitations of this approach is that the proportion of neurons differentiated from UCB-MSCs still remains at low level. Thus, to induce UCB-MSCs to differentiate into neuron-like cells with a higher proportion is one of the key technologies of regenerative medicine and tissue engineering. Many induction protocols with remarkably higher differentiation rate to neurons have been reported. However, each protocol has its pros and cons and whether the neurons differentiated from UCB-MSCs under a certain protocol has normal nerve function remains controversial. Therefore, to guarantee the success of future clinical applications of UCB-MSCs, more investigations should be performed to improve the induction method and differentiation efficiency.
Collapse
Affiliation(s)
- Lei Wang
- 1Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA) , Changsha, Hunan , China
| | | |
Collapse
|
12
|
Kim HO, Choi SM, Kim HS. Mesenchymal stem cell-derived secretome and microvesicles as a cell-free therapeutics for neurodegenerative disorders. Tissue Eng Regen Med 2013. [DOI: 10.1007/s13770-013-0010-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
13
|
Catacchio I, Berardi S, Reale A, De Luisi A, Racanelli V, Vacca A, Ria R. Evidence for bone marrow adult stem cell plasticity: properties, molecular mechanisms, negative aspects, and clinical applications of hematopoietic and mesenchymal stem cells transdifferentiation. Stem Cells Int 2013; 2013:589139. [PMID: 23606860 PMCID: PMC3625599 DOI: 10.1155/2013/589139] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/22/2013] [Indexed: 12/24/2022] Open
Abstract
In contrast to the pluripotent embryonic stem cells (ESCs) which are able to give rise to all cell types of the body, mammalian adult stem cells (ASCs) appeared to be more limited in their differentiation potential and to be committed to their tissue of origin. Recently, surprising new findings have contradicted central dogmas of commitment of ASCs by showing their plasticity to differentiate across tissue lineage boundaries, irrespective of classical germ layer designations. The present paper supports the plasticity of the bone marrow stem cells (BMSCs), bringing the most striking and the latest evidences of the transdifferentiation properties of the bone marrow hematopoietic and mesenchymal stem cells (BMHSCs, and BMMSCs), the two BM populations of ASCs better characterized. In addition, we report the possible mechanisms that may explain these events, outlining the clinical importance of these phenomena and the relative problems.
Collapse
Affiliation(s)
- Ivana Catacchio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Simona Berardi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Antonia Reale
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Annunziata De Luisi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| |
Collapse
|
14
|
Kim JW, Lee JH, Lyoo YS, Jung DI, Park HM. The effects of topical mesenchymal stem cell transplantation in canine experimental cutaneous wounds. Vet Dermatol 2013; 24:242-e53. [PMID: 23432413 PMCID: PMC3618380 DOI: 10.1111/vde.12011] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2012] [Indexed: 12/26/2022]
Abstract
Background Adult stem cells have been widely investigated in bioengineering approaches for tissue repair therapy. We evaluated the clinical value and safety of the application of cultured bone marrow-derived allogenic mesenchymal stem cells (MSCs) for treating skin wounds in a canine model. Hypothesis Topical allogenic MSC transplantation can accelerate the closure of experimental full-thickness cutaneous wounds and attenuate local inflammation. Animals Adult healthy beagle dogs (n = 10; 3–6 years old; 7.2–13.1 kg) were studied. Methods Full-thickness skin wounds were created on the dorsum of healthy beagles, and allogenic MSCs were injected intradermally. The rate of wound closure and the degree of collagen production were analysed histologically using haematoxylin and eosin staining and trichrome staining. The degree of cellular proliferation and angiogenesis was evaluated by immunocytochemistry using proliferating cell nuclear antigen-, vimentin- and α-smooth muscle actin-specific antibodies. Local mRNA expression levels of interleukin-2, interferon-γ, basic fibroblast growth factor and matrix metalloproteinase-2 were evaluated by RT-PCR. Results Compared with the vehicle-treated wounds, MSC-treated wounds showed more rapid wound closure and increased collagen synthesis, cellular proliferation and angiogenesis. Moreover, MSC-treated wounds showed decreased expression of pro-inflammatory cytokines (interleukin-2 and interferon-γ) and wound healing-related factors (basic fibroblast growth factor and matrix metalloproteinase-2). Conclusion and clinical importance Topical transplantation of MSCs results in paracrine effects on cellular proliferation and angiogenesis, as well as modulation of local mRNA expression of several factors related to cutaneous wound healing. Résumé Resumen Zusammenfassung 摘要 要約
Collapse
Affiliation(s)
- Ju-Won Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | | | | | | | | |
Collapse
|
15
|
Electrophysiological properties and synaptic function of mesenchymal stem cells during neurogenic differentiation - a mini-review. Int J Artif Organs 2012; 35:323-37. [PMID: 22505200 DOI: 10.5301/ijao.5000085] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2011] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) have gained considerable interest due to their potential use in cell therapies and tissue engineering. They have been reported to differentiate into various anchorage-dependent cell types, including bone, cartilage, and tendon. Our focus is on the differentiation of MSCs into neuron-like cells through the use of soluble chemical stimuli or specific growth factor supplements. The resulting cells appear to adopt neural phenotypes and express some typical neuronal markers, however, their electrophysiological properties and synaptic function remains unclear. RESULTS This mini-review illustrates how particular characteristics, electrophysiological properties, and synaptic functions of MSCs change during their neuronal differentiation. In particular we focus on changes in ion currents, ion channels, synaptic communication, and neurotransmitter release. We also highlight conflicting results, caused by inconsistencies in the experimental conditions used and in the methodologies adopted. CONCLUSIONS We conclude that there is insufficient data and that further, carefully controlled investigations are required in order to ascertain whether MSC-derived neuron-like cells can exhibit the necessary neuronal functions to become clinically relevant for use in neural repairs.
Collapse
|
16
|
Neural crest stem cells from dental tissues: a new hope for dental and neural regeneration. Stem Cells Int 2012; 2012:103503. [PMID: 23093977 PMCID: PMC3472918 DOI: 10.1155/2012/103503] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 08/13/2012] [Accepted: 09/05/2012] [Indexed: 12/12/2022] Open
Abstract
Several stem cell sources persist in the adult human body, which opens the doors to both allogeneic and autologous cell therapies. Tooth tissues have proven to be a surprisingly rich and accessible source of neural crest-derived ectomesenchymal stem cells (EMSCs), which may be employed to repair disease-affected oral tissues in advanced regenerative dentistry. Additionally, one area of medicine that demands intensive research on new sources of stem cells is nervous system regeneration, since this constitutes a therapeutic hope for patients affected by highly invalidating conditions such as spinal cord injury, stroke, or neurodegenerative diseases. However, endogenous adult sources of neural stem cells present major drawbacks, such as their scarcity and complicated obtention. In this context, EMSCs from dental tissues emerge as good alternative candidates, since they are preserved in adult human individuals, and retain both high proliferation ability and a neural-like phenotype in vitro. In this paper, we discuss some important aspects of tissue regeneration by cell therapy and point out some advantages that EMSCs provide for dental and neural regeneration. We will finally review some of the latest research featuring experimental approaches and benefits of dental stem cell therapy.
Collapse
|
17
|
Faça VM. Human mesenchymal stromal cell proteomics: contribution for identification of new markers and targets for medicine intervention. Expert Rev Proteomics 2012; 9:217-30. [PMID: 22462791 DOI: 10.1586/epr.12.9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs) have become of great interest for cell-based therapy owing to their roles in tissue repair and immune suppression. MSCs have the ability to differentiate into specialized tissues, including bone, cartilage and muscle, among several others. Furthermore, it has been found that MSCs can also serve as cellular factories that secrete mediators to stimulate in situ regeneration of injured tissues. Proteomics has contributed significantly to the identification of new proteins to improve cellular characterization of MSCs, to identify new targets for therapeutic intervention and to elucidate important pathways utilized by MSCs to differentiate into distinct tissues. As proteomics technology advances, several studies can be revisited and analyzed in depth, employing state-of-the-art approaches, helping to uncover the cellular mechanisms utilized by MSCs to exert their regenerative functionalities. In this article, we will review the progress made so far and discuss further opportunities for proteomics to contribute to the clinical applications of MSCs.
Collapse
Affiliation(s)
- Vitor Marcel Faça
- Department of Biochemistry & Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Brazil.
| |
Collapse
|
18
|
Dalous J, Larghero J, Baud O. Transplantation of umbilical cord-derived mesenchymal stem cells as a novel strategy to protect the central nervous system: technical aspects, preclinical studies, and clinical perspectives. Pediatr Res 2012; 71:482-90. [PMID: 22430384 DOI: 10.1038/pr.2011.67] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The prevention of perinatal neurological disabilities remains a major challenge for public health, and no neuroprotective treatment to date has proven clinically useful in reducing the lesions leading to these disabilities. Efforts are, therefore, urgently needed to test other neuroprotective strategies including cell therapies. Although stem cells have raised great hopes as an inexhaustible source of therapeutic products that could be used for neuroprotection and neuroregeneration in disorders affecting the brain and spinal cord, certain sources of stem cells are associated with potential ethical issues. The human umbilical cord (hUC) is a rich source of stem and progenitor cells including mesenchymal stem cells (MSCs) derived either from the cord or from cord blood. hUC MSCs (hUC-MSCs) have several advantages as compared to other types and sources of stem cells. In this review, we will summarize the most recent findings regarding the technical aspects and the preclinical investigation of these promising cells in neuroprotection and neuroregeneration, and their potential use in the developing human brain. However, extensive studies are needed to optimize the administration protocol, safety parameters, and potential preinjection cell manipulations before designing a controlled trial in human neonates.
Collapse
Affiliation(s)
- Jérémie Dalous
- INSERM UMR 676, Université Paris Diderot, Hôpital Robert Debré, APHP, Paris, France
| | | | | |
Collapse
|
19
|
Liu H, Yang K, Xin T, Wu W, Chen Y. Implanted electro-acupuncture electric stimulation improves outcome of stem cells' transplantation in spinal cord injury. ACTA ACUST UNITED AC 2012; 40:331-7. [PMID: 22384853 DOI: 10.3109/10731199.2012.659350] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Spinal cord injury (SCI) is one of the most serious disorders in clinics, and the high disability rate and functional deficits are common issues in patients. Transplantation of bone-marrow-derived mesenchymal stromal cells (BMSCs) into the injured spinal cord is emerging as a novel method in the therapeutics of SCI; however, its application is limited by the poor survival rate of the transplanted cells and low differentiation rate into neurons. Our laboratory recently reported that electrical stimulation (ES) dramatically improves the survival rate of transplanted BMSCs and increases spinal cord functions in animals with spinal cord injury. In this paper, we asked whether implanted electro-acupuncture (iEA) can advance the beneficial effects from the ES treatment in animals with spinal cord injury. We showed that BMSCs transplantation alone resulted in significant functional recovery in animals. Interestingly, iEA with BMSCs treatment induced a significantly higher functional improvement in locomotor functions and SSEP compared to the BMSCs treatment alone. Additionally, we used molecular biology techniques and showed that BMSCs transplantation with iEA treatment significantly increased the number of surviving BMSCs compared to the BMSCs alone group. In conclusion, our experiment showed that the approach of coupling iEA electric stimulation and BMSCs transplantation remarkably promotes functional improvements in animals with spinal cord injury and holds promising potential to treat spinal cord injury in humans.
Collapse
Affiliation(s)
- Haichun Liu
- Department of Orthopaedic and Trauma Surgery, Shandong University Qilu Hospital, P. R. China
| | | | | | | | | |
Collapse
|
20
|
Soleimani M, Abbasnia E, Fathi M, Sahraei H, Fathi Y, Kaka G. The effects of low-level laser irradiation on differentiation and proliferation of human bone marrow mesenchymal stem cells into neurons and osteoblasts--an in vitro study. Lasers Med Sci 2012; 27:423-430. [PMID: 21597948 DOI: 10.1007/s10103-011-0930-1] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 04/18/2011] [Indexed: 12/12/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) are promising for use in regenerative medicine. Several studies have shown that low-level laser irradiation (LLLI) could affect the differentiation and proliferation of MSCs. The aim of this study was to examine the influence of LLLI at different energy densities on BMSCs differentiation into neuron and osteoblast. Human BMSCs were cultured and induced to differentiate to either neuron or osteoblast in the absence or presence of LLLI. Gallium aluminum arsenide (GaAlAs) laser irradiation (810 nm) was applied at days 1, 3, and 5 of differentiation process at energy densities of 3 or 6 J/cm(2) for BMSCs being induced to neurons, and 2 or 4 J/cm(2) for BMSCs being induced to osteoblasts. BMSCs proliferation was evaluated by MTT assay on the seventh day of differentiation. BMSCs differentiation to neurons was assessed by immunocytochemical analysis of neuron-specific enolase on the seventh day of differentiation. BMSCs differentiation to osteoblast was tested on the second, fifth, seventh, and tenth day of differentiation via analysis of alkaline phosphatase (ALP) activity. LLLI promoted BMSCs proliferation significantly at all energy densities except for 6 J/cm(2) in comparison to control groups on the seventh day of differentiation. LLLI at energy densities of 3 and 6 J/cm(2) dramatically facilitated the differentiation of BMSCs into neurons (p < 0.001). Also, ALP activity was significantly enhanced in irradiated BMSCs differentiated to osteoblast on the second, fifth, seventh, and tenth day of differentiation (p < 0.001 except for the second day). Using LLLI at 810 nm wavelength enhances BMSCs differentiation into neuron and osteoblast in the range of 2-6 J/cm(2), and at the same time increases BMSCs proliferation (except for 6 J/cm(2)). The effect of LLLI on differentiation and proliferation of BMSCs is dose-dependent. Considering these findings, LLLI could improve current in vitro methods of differentiating BMSCs prior to transplantation.
Collapse
Affiliation(s)
- Masoud Soleimani
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| | | | | | | | | | | |
Collapse
|
21
|
Titomanlio L, Kavelaars A, Dalous J, Mani S, El Ghouzzi V, Heijnen C, Baud O, Gressens P. Stem cell therapy for neonatal brain injury: perspectives and challenges. Ann Neurol 2012; 70:698-712. [PMID: 22162055 DOI: 10.1002/ana.22518] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebral palsy is a major health problem caused by brain damage during pregnancy, delivery, or the immediate postnatal period. Perinatal stroke, intraventricular hemorrhage, and asphyxia are the most common causes of neonatal brain damage. Periventricular white matter damage (periventricular leukomalacia) is the predominant form in premature infants and the most common antecedent of cerebral palsy. Stem cell treatment has proven effective in restoring injured organs and tissues in animal models. The potential of stem cells for self-renewal and differentiation translates into substantial neuroprotection and neuroregeneration in the animal brain, with minimal risks of rejection and side effects. Stem cell treatments described to date have used neural stem cells, embryonic stem cells, mesenchymal stem cells, umbilical cord stem cells, and induced pluripotent stem cells. Most of these treatments are still experimental. In this review, we focus on the efficacy of stem cell therapy in animal models of cerebral palsy, and discuss potential implications for current and future clinical trials.
Collapse
|
22
|
Comparison of long-term retinoic acid-based neural induction methods of bone marrow human mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2011; 47:484-91. [PMID: 21638161 DOI: 10.1007/s11626-011-9425-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 05/11/2011] [Indexed: 12/12/2022]
Abstract
The differentiation of human mesenchymal stem cells (hMSCs) into neural cells in vitro provides a potential tool to be utilized for cell therapy of neurodegenerative disorders. Although previous studies repeated different protocols for the induction of neural cells from hMSCs in vitro, the results were not in complete agreement. In this study, we have attempted to compare three of these neural induction methods; retinoic acid (RA) treatment, RA treatment in serum reduced conditions, and treatment using other chemical compounds (dimethyl sulfoxide and potassium chloride) along with RA by real-time cell analysis and immunofluorescent staining of neural markers. RA treatment led to a slow progression of cells into neural-like morphology with the expression of neural protein neurofilament whereas reducing serum during RA treatment caused a much more extended differentiation process. Additionally, neural-like morphology was persistent in the later periods of differentiation in RA treatment. On the other hand, chemical induction caused cell shrinkages mimicking neural-like morphology in a short time and loss of this morphology along with increased cell death in later periods. Among the three methods compared, RA treatment was the most reliable one in terms of stability of differentiation and neural protein expressions.
Collapse
|
23
|
Lee JH, Yu HS, Lee GS, Ji A, Hyun JK, Kim HW. Collagen gel three-dimensional matrices combined with adhesive proteins stimulate neuronal differentiation of mesenchymal stem cells. J R Soc Interface 2011; 8:998-1010. [PMID: 21247946 DOI: 10.1098/rsif.2010.0613] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional gel matrices provide specialized microenvironments that mimic native tissues and enable stem cells to grow and differentiate into specific cell types. Here, we show that collagen three-dimensional gel matrices prepared in combination with adhesive proteins, such as fibronectin (FN) and laminin (LN), provide significant cues to the differentiation into neuronal lineage of mesenchymal stem cells (MSCs) derived from rat bone marrow. When cultured within either a three-dimensional collagen gel alone or one containing either FN or LN, and free of nerve growth factor (NGF), the MSCs showed the development of numerous neurite outgrowths. These were, however, not readily observed in two-dimensional culture without the use of NGF. Immunofluorescence staining, western blot and fluorescence-activated cell sorting analyses demonstrated that a large population of cells was positive for NeuN and glial fibrillary acidic protein, which are specific to neuronal cells, when cultured in the three-dimensional collagen gel. The dependence of the neuronal differentiation of MSCs on the adhesive proteins containing three-dimensional gel matrices is considered to be closely related to focal adhesion kinase (FAK) activation through integrin receptor binding, as revealed by an experiment showing no neuronal outgrowth in the FAK-knockdown cells and stimulation of integrin β1 gene. The results provided herein suggest the potential role of three-dimensional collagen-based gel matrices combined with adhesive proteins in the neuronal differentiation of MSCs, even without the use of chemical differentiation factors. Furthermore, these findings suggest that three-dimensional gel matrices might be useful as nerve-regenerative scaffolds.
Collapse
Affiliation(s)
- Jae Ho Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, , South Korea
| | | | | | | | | | | |
Collapse
|
24
|
Wu W, Zhao H, Xie B, Liu H, Chen Y, Jiao G, Wang H. Implanted spike wave electric stimulation promotes survival of the bone marrow mesenchymal stem cells and functional recovery in the spinal cord injured rats. Neurosci Lett 2011; 491:73-8. [PMID: 21232582 DOI: 10.1016/j.neulet.2011.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/27/2010] [Accepted: 01/04/2011] [Indexed: 12/20/2022]
Abstract
Transplantation of bone marrow-derived mesenchymal stromal cells (BMSCs) into the injured spinal cord may provide therapeutic benefit, but its application is limited by their poor survival and low differentiation rate into neurons. Electrical stimulation (ES) has been reported to promote survival and differentiation of the BMSCs. Therefore we investigated whether implanted spike wave ES could improve survival of BMSCs after transplantation and result in functional improvement in animals with spinal cord injury. Our results showed that the number and ratio of survived BMSCs near the lesion site were significantly increased in the BMSCs+ES-treated group as compared to BMSCs transplantation or ES treatment alone group. Furthermore, results from BBB scales, SSEP and DTI demonstrated a significant improved functional recovery in the BMSCs+ES group. This indicated that implanted spike wave ES could promote the bioactivity of BMSCs and their survival. This represents a new therapeutic potential of the combination of BMSCs transplantation with implanted spike wave ES to treat spinal cord injury.
Collapse
Affiliation(s)
- Wenliang Wu
- Department of Trauma Surgery, Shandong University Qilu Hospital, Wenhuaxi Road 107, Jinan 250012, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Fox LE, Shen J, Ma K, Liu Q, Shi G, Pappas GD, Qu T, Cheng J. Membrane properties of neuron-like cells generated from adult human bone-marrow-derived mesenchymal stem cells. Stem Cells Dev 2010; 19:1831-41. [PMID: 20394468 PMCID: PMC3128307 DOI: 10.1089/scd.2010.0089] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Accepted: 04/15/2010] [Indexed: 11/12/2022] Open
Abstract
Adult mesenchymal stem cells (MeSCs) isolated from human bone marrow are capable of generating neural stem cell (NSC)-like cells that can be subsequently differentiated into cells expressing molecular markers for neurons. Here we report that these neuron-like cells had functional properties similar to those of brain-derived neurons. Whole-cell patch-clamp recordings and calcium imaging experiments were performed on neuron-like cells differentiated from bone-marrow-derived NSC-like cells. The neuron-like cells were subjected to current pulses to determine if they were capable of generating depolarization-induced action potentials. We found that nearly all of the cells with neuron-like morphology exhibited active membrane properties in response to the depolarizing pulses. The most common response was a single spike-like event with an overshoot and brief afterhyperpolarization. Cells that did not generate overshooting spike-like events usually displayed rectifying current-voltage relationships. The prevalence of these active membrane properties in response to the depolarizing current pulses suggested that the human MeSCs (hMeSCs) were capable of converting to a neural lineage under defined culture conditions. The spike-like events were blocked by the voltage-gated sodium channel inhibitor lidocaine, but unaffected by another sodium channel inhibitor tetrodotoxin (TTX). In calcium imaging experiments, the neuron-like cells responded to potassium chloride depolarization and l-glutamate application with increases in the cytoplasmic calcium levels. Thus, the neuron-like cells appeared to express TTX-resistant voltage-gated sodium channels, voltage-gated calcium channels, and functional l-glutamate receptors. These results demonstrate that hMeSCs were capable of generating cells with characteristics typical of functional neurons that may prove useful for neuroreplacement therapies.
Collapse
Affiliation(s)
- Lyle E. Fox
- Department of Pain Management, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Jun Shen
- Department of Pain Management, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Ke Ma
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois at Chicago (UIC), Chicago, Illinois
| | - Qing Liu
- Department of Pain Management, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Guangbin Shi
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois at Chicago (UIC), Chicago, Illinois
| | - George D. Pappas
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois at Chicago (UIC), Chicago, Illinois
| | - Tingyu Qu
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois at Chicago (UIC), Chicago, Illinois
| | - Jianguo Cheng
- Department of Pain Management, Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|
26
|
Lepski G, Jannes CE, Strauss B, Marie SK, Nikkhah G. Survival and Neuronal Differentiation of Mesenchymal Stem Cells Transplanted into the Rodent Brain Are Dependent upon Microenvironment. Tissue Eng Part A 2010; 16:2769-82. [DOI: 10.1089/ten.tea.2009.0686] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Guilherme Lepski
- Department of Stereotactic and Functional Neurosurgery, Albert Ludwigs University, Frieburg, Germany
| | - Cinthia E. Jannes
- Laboratory of Molecular Biology LIM15, Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Bryan Strauss
- Laboratory of Molecular Biology, INCOR, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Suely K.N. Marie
- Laboratory of Molecular Biology LIM15, Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Guido Nikkhah
- Department of Stereotactic and Functional Neurosurgery, Neurocentre, Albert-Ludwig University, Freiburg, Germany
| |
Collapse
|
27
|
Cholinergic neuronal differentiation of bone marrow mesenchymal stem cells in rhesus monkeys. SCIENCE CHINA-LIFE SCIENCES 2010; 53:573-80. [PMID: 20596940 DOI: 10.1007/s11427-010-0009-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2009] [Accepted: 09/29/2009] [Indexed: 10/19/2022]
Abstract
The purpose of the present study was to determine the best cholinergic neuronal differentiation method of rhesus monkey bone marrow mesenchymal stem cells (BMSCs). Four methods were used to induce differentiation, and the groups were assigned accordingly: basal inducing group (culture media, bFGF, and forskolin); SHH inducing group (SHH, inducing group); RA inducing group (RA, basal inducing group); and SHH+RA inducing group (SHH, RA, and basal inducing group). All groups displayed neuronal morphology and increased expression of nestin and neuron-specific enolase. The basal inducing group did not express synapsin, and cells from the SHH inducing group did not exhibit neuronal resting membrane potential. In contrast, results demonstrated that BMSCs from the RA and SHH+RA inducing groups exhibited neuronal resting membrane potential, and cells from the SHH+RA inducing group expressed higher levels of synapsin and acetylcholine. In conclusion, the induction of cholinergic differentiation through SHH+RA was determined to be superior to the other methods.
Collapse
|
28
|
Sadan O, Melamed E, Offen D. Bone-marrow-derived mesenchymal stem cell therapy for neurodegenerative diseases. Expert Opin Biol Ther 2010; 9:1487-97. [PMID: 19821796 DOI: 10.1517/14712590903321439] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Stem-cell-based therapy is a promising new approach to handling neurodegenerative diseases. One of the most promising cellular sources is bone-marrow-derived mesenchymal stem cells (MSCs) also termed multipotent stromal cells. MSCs represent an autologous source and are abundant and non-tumorigenic. Additionally, MSCs possess the useful characteristics of homing and chemokine secretion. OBJECTIVE/METHODS Since neurodegenerative diseases have many pathological processes in common, a specific therapeutic agent could potentially ameliorate the symptoms of several distinct neurodegenerative diseases. In this review we demonstrate the wide variety of mechanisms by which MSCs can influence neurodegenerative processes. RESULTS/CONCLUSIONS The mechanisms by which transplanted MSCs influence neurodegenerative diseases can be broadly classified as cellular replacement or paracrine secretion, with the latter subdivided into trophic factor secretion or immunomodulation by cytokines. Emerging research suggests that genetic manipulations before transplantation could enhance the therapeutic potential of MSCs. Such manipulation could turn the cells into a 'Trojan horse', to deliver specific proteins, or promote reprogramming of the MSCs into the neural lineage. Clinical trials testing MSC-based therapies for familial amyotrophic lateral sclerosis and multiple sclerosis are in progress.
Collapse
Affiliation(s)
- Ofer Sadan
- Neurosciences Laboratory, Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | | | | |
Collapse
|
29
|
Studenovská H, Vodička P, Proks V, Hlučilová J, Motlík J, Rypáček F. Synthetic poly(amino acid) hydrogels with incorporated cell-adhesion peptides for tissue engineering. J Tissue Eng Regen Med 2010; 4:454-63. [DOI: 10.1002/term.256] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
30
|
Lepski G, Jannes CE, Maciaczyk J, Papazoglou A, Mehlhorn AT, Kaiser S, Teixeira MJ, Marie SK, Bischofberger J, Nikkhah G. Limited Ca2+ and PKA-pathway dependent neurogenic differentiation of human adult mesenchymal stem cells as compared to fetal neuronal stem cells. Exp Cell Res 2010; 316:216-31. [DOI: 10.1016/j.yexcr.2009.08.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 08/03/2009] [Accepted: 08/11/2009] [Indexed: 11/16/2022]
|
31
|
Lee SK, Kim Y, Kim SS, Lee JH, Cho K, Lee SS, Lee ZW, Kwon KH, Kim YH, Suh-Kim H, Yoo JS, Park YM. Differential expression of cell surface proteins in human bone marrow mesenchymal stem cells cultured with or without basic fibroblast growth factor containing medium. Proteomics 2009; 9:4389-405. [PMID: 19655310 DOI: 10.1002/pmic.200900165] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells, which have the capability to differentiate into various mesenchymal tissues such as bone, cartilage, fat, tendon, muscle, and marrow stroma. However, they lose the capability of multi-lineage differentiation after several passages. It is known that basic fibroblast growth factor (bFGF) increases growth rate, differentiation potential, and morphological changes of MSCs in vitro. In this report, we have used 2-DE coupled to MS to identify differentially expressed proteins at the cell membrane level in MSCs growing in bFGF containing medium. The cell surface proteins isolated by the biotin-avidin affinity column were separated by 2-DE in triplicate experiments. A total of 15 differentially expressed proteins were identified by quadrupole-time of flight tandem MS. Nine of the proteins were upregulated and six proteins were downregulated in the MSCs cultured with bFGF containing medium. The expression level of three actin-related proteins, F-actin-capping protein subunit alpha-1, actin-related protein 2/3 complex subunit 2, and myosin regulatory light chain 2, was confirmed by Western blot analysis. The results indicate that the expression levels of F-actin-capping protein subunit alpha-1, actin-related protein 2/3 complex subunit 2, and myosin regulatory light chain 2 are important in bFGF-induced morphological change of MSCs.
Collapse
Affiliation(s)
- Sang Kwang Lee
- Mass Spectrometry Research Center, Korea Basic Science Institute, Daejeon, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chao YX, He BP, Tay SSW. Mesenchymal stem cell transplantation attenuates blood brain barrier damage and neuroinflammation and protects dopaminergic neurons against MPTP toxicity in the substantia nigra in a model of Parkinson's disease. J Neuroimmunol 2009; 216:39-50. [PMID: 19819031 DOI: 10.1016/j.jneuroim.2009.09.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Revised: 09/03/2009] [Accepted: 09/04/2009] [Indexed: 01/14/2023]
Abstract
Immunomodulatory effects of transplanted mesenchymal stem cells (MSCs) in the treatment of Parkinson's disease were studied in the MPTP-induced mouse model. MPTP treatment induced a significant loss of dopaminergic neurons, decreased expressions of claudin 1, claudin 5 and occludin in the substantia nigra compacta (SNc), and functional damage of the blood brain barrier (BBB). Our study further discovered that infiltration of MBLs into the brain to bind with microglia was detected in the SNc of MPTP-treated mice, suggesting that the BBB compromise and MBL infiltration might be involved in the pathogenesis of MPTP-induced PD. In addition, MPTP treatment also increased the expression of mannose-binding lectins (MBLs) in the liver tissue. Intravenous transplantation of MSCs into MPTP-treated mice led to recovery of BBB integrity, suppression of MBL infiltration at SNc and MBL expression in the liver, suppression of microglial activation and prevention of dopaminergic neuron death. No transplanted MSCs were observed to differentiate into dopaminergic neurons, while the MSCs migrated into the SNc and released TGF-beta1 there. Therefore, intravenous transplantation of MSCs which protect dopaminergic neurons from MPTP toxicity may be engaged in anyone or a combination of these mechanisms: repair of the BBB, reduction of MBL in the brain, inhibition of microglial cytotoxicity, and direct protection of dopaminergic neurons.
Collapse
Affiliation(s)
- Yin Xia Chao
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
33
|
Kuo YC, Yeh CF, Yang JT. Differentiation of bone marrow stromal cells in poly(lactide-co-glycolide)/chitosan scaffolds. Biomaterials 2009; 30:6604-13. [PMID: 19712972 DOI: 10.1016/j.biomaterials.2009.08.028] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 08/13/2009] [Indexed: 10/20/2022]
Abstract
This study investigates the physicochemical properties of poly(lactide-co-glycolide) (PLGA)/chitosan scaffolds and the neuron growth factor (NGF)-guided differentiation of bone marrow stromal cells (BMSCs) in the scaffolds. The scaffolds were prepared by the crosslinking of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N-hydroxysuccinimide and genipin, and the differentiating BMSCs were characterized against CD44, CD90 and NeuN. The scaffold with 20% PLGA yielded 95% porosity, Young's modulus of 13MPa, 70% adhesion of BMSCs and 1.6-fold increase in the cell viability over 7-day cultivation. BMSCs without guidance in the PLGA/chitosan scaffolds were prone to differentiate toward osteoblasts with apparent deposition of calcium. When NGF was introduced, an increased weight percentage of PLGA yielded more identified neurons. In addition, mature neurons emerged from the PLGA-rich biomaterials after induction with NGF over 2 days. A proper control over the physical and biomedical property of the scaffolds and the NGF-guided differentiation of BMSCs can be promising for nerve regeneration.
Collapse
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, Republic of China.
| | | | | |
Collapse
|
34
|
Cho YM, Jang YS, Jang YM, Chung SM, Kim HS, Lee JH, Jeong SW, Kim IK, Kim JJ, Kim KS, Kwon OJ. Induction of unfolded protein response during neuronal induction of rat bone marrow stromal cells and mouse embryonic stem cells. Exp Mol Med 2009; 41:440-52. [PMID: 19322020 DOI: 10.3858/emm.2009.41.6.049] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
When we treated rat bone marrow stromal cells (rBMSCs) with neuronal differentiation induction media, typical unfolded protein response (UPR) was observed. BIP/GRP78 protein expression was time-dependently increased, and three branches of UPR were all activated. ATF6 increased the transcription of XBP1 which was successfully spliced by IRE1. PERK was phosphorylated and it was followed by eIF2alpha phosphorylation. Transcription of two downstream targets of eIF2alpha, ATF4 and CHOP/GADD153, were transiently up-regulated with the peak level at 24 h. Immunocytochemical study showed clear coexpression of BIP and ATF4 with NeuN and Map2, respectively. UPR was also observed during the neuronal differentiation of mouse embryonic stem (mES) cells. Finally, chemical endoplasmic reticulum (ER) stress inducers, thapsigargin, tunicamycin, and brefeldin A, dose-dependently increased both mRNA and protein expressions of NF-L, and, its expression was specific to BIP-positive rBMSCs. Our results showing the induction of UPR during neuronal differentiations of rBMSCs and mES cells as well as NF-L expression by ER stress inducers strongly suggest the potential role of UPR in neuronal differentiation.
Collapse
Affiliation(s)
- Yoon Mi Cho
- Department of Biochemistry, 2MRC for Cell Death Disease Research Center, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Shetty P, Ravindran G, Sarang S, Thakur AM, Rao HS, Viswanathan C. Clinical grade mesenchymal stem cells transdifferentiated under xenofree conditions alleviates motor deficiencies in a rat model of Parkinson's disease. Cell Biol Int 2009; 33:830-8. [PMID: 19465139 DOI: 10.1016/j.cellbi.2009.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 01/13/2009] [Accepted: 05/07/2009] [Indexed: 12/29/2022]
Abstract
UNLABELLED Bone marrow derived mesenchymal stem cells (BMMSCs) is a valid, definitive candidate for repair of damaged tissues in degenerative disorders in general and neurological diseases in particular. We have standardized the processing conditions for proliferation of BMMSCs using xenofree medium and checked their in vitro and in vivo neurogenic potential. METHOD The proliferative potential of BMMSCs was analyzed using xenofree media and functionality checked by transplantation into Parkinson's disease (PD) animal models. In vitro neuronal differentiation was investigated by neuronal induction media supplemented with growth factors. Differentiated cells were characterized at cellular and molecular levels. In vitro functionality estimated by dopamine secretion. RESULTS A pure population of BMMSCs showing an 8-10 fold expansion was obtained using xenofree media. On differentiation to neuronal lineage, they exhibited neuronal morphology. Detectable levels of dopamine (1.93 ng/ml) were secreted into the culture media of differentiated cells. There was a significant behavioural improvement in PD models 3 months post transplantation. CONCLUSION Our study demonstrates that BMMSCs can be transdifferentiated efficiently into functional dopaminergic neurons both in vitro and in vivo. This holds immense clinical potential as a replacement therapy for PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Prathibha Shetty
- Regenerative Medicine, Haematopoietic Stem Cell Group, Dhirubhai Ambani Life Sciences Centre, R-282 TTC Area of MIDC, Thane Belapur Road, Rabale, Navi Mumbai 400701, India.
| | | | | | | | | | | |
Collapse
|
36
|
Wu L, Wei X, Ling J, Liu L, Liu S, Li M, Xiao Y. Early osteogenic differential protein profile detected by proteomic analysis in human periodontal ligament cells. J Periodontal Res 2009; 44:645-56. [PMID: 19453858 DOI: 10.1111/j.1600-0765.2008.01174.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Human periodontal ligament cells play a pivotal role in maintaining periodontal ligament space, contain progenitors that are able to differentiate into cementoblasts/osteoblasts and have a tremendous potential to regenerate periodontal tissue. However, the exact molecular mechanisms governing the differentiation mechanisms of progenitors in periodontal ligament cells remain largely unknown. This study was carried out to investigate the differentially expressed proteins involved in the osteogenic differentiation of progenitors presented in periodontal ligament cells. MATERIAL AND METHODS Using two-dimensional gel electrophoresis, mass spectrometry and peptide mass fingerprinting, we analyzed the differential protein profiles of periodontal ligament cells undergoing mineralization. RESULTS Compared with undifferentiated periodontal ligament cells, 61 proteins in periodontal ligament cells undergoing differentiation showed at least a 1.5-fold change in intensity, of which 29 differentially expressed proteins were successfully identified by matrix-assisted laser-desorption ionization time-of-flight mass spectrometry. The expression of some of the identified proteins was further confirmed by western blotting and reverse transcription-polymerase chain reaction analysis. The identified proteins were cytoskeleton proteins and cytoskeleton-associated proteins, nuclear proteins and cell membrane-bound molecules. CONCLUSION Our results suggest that the proteins identified in this study may be associated with the unique function of periodontal ligament cells in maintaining periodontal tissue homeostasis, thus providing a comprehensive reference for understanding and investigating in greater detail the molecular mechanisms of periodontal ligament cells involved in periodontal regeneration.
Collapse
Affiliation(s)
- L Wu
- Department of Orthodontics, Guanghua School of Stomatology, Sun Yat-Sen University, Guangdong, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Chae SW, Jee BK, Lee JY, Han CW, Jeon YW, Lim Y, Lee KH, Rha HK, Chae GT. HOX gene analysis in the osteogenic differentiation of human mesenchymal stem cells. Genet Mol Biol 2008. [DOI: 10.1590/s1415-47572008005000019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Song Wha Chae
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Bo Keun Jee
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Joo Yong Lee
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Chang Whan Han
- Department of Orthopedic Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Yang-Whan Jeon
- Department of Psychiatry, Our Lady of Mercy Hospital, The Catholic University of Korea, Republic of Korea
| | - Young Lim
- Department of Occupational and Environmental Medicine, St. Mary's Hospital, The Catholic University of Korea, Republic of Korea
| | - Kweon-Haeng Lee
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Hyoung Kyun Rha
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Gue-Tae Chae
- Institute of Hansen's Disease, The Catholic University of Korea, Republic of Korea
| |
Collapse
|
38
|
Zwart I, Hill AJ, Girdlestone J, Manca MF, Navarrete R, Navarrete C, Jen LS. Analysis of neural potential of human umbilical cord blood-derived multipotent mesenchymal stem cells in response to a range of neurogenic stimuli. J Neurosci Res 2008; 86:1902-15. [PMID: 18338797 DOI: 10.1002/jnr.21649] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We investigated the neurogenic potential of full-term human umbilical cord blood (hUCB)-derived multipotent mesenchymal stem cells (MSCs) in response to neural induction media or coculture with rat neural cells. Phenotypic and functional changes were assessed by immunocytochemistry, RT-PCR, and whole-cell patch-clamp recordings. Naive MSCs expressed both mesodermal and ectodermal markers prior to neural induction. Exposure to retinoic acid, basic fibroblast growth factor, or cyclic adenosine monophosphate (cAMP) did not stimulate neural morphology, whereas exposure to dibutyryl cAMP and 3-isobutyl-1-methylxanthine stimulated a neuron-like morphology but also appeared to be cytotoxic. All protocols stimulated increases in expression of the neural precursor marker nestin, but expression of mature neuronal or glial markers MAP2 and GFAP was not observed. Nestin expression increases were serum level dependent. Electrophysiological properties of MSCs were studied with whole-cell patch-clamp recordings. The MSCs possessed no ionic currents typical of neurons before or after neural induction protocols. Coculture of hUCB-derived MSCs and rat neural cells induced some MSCs to adopt an astrocyte-like morphology and express GFAP protein and mRNA. Our data suggest hUCB-derived MSCs do not transdifferentiate into mature functioning neurons in response to the above neurogenic protocols; however, coculture with rat neural cells led to a minority adopting an astrocyte-like phenotype.
Collapse
Affiliation(s)
- Isabel Zwart
- Department of Cellular and Molecular neuroscience, Imperial College, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
39
|
Breunig JJ, Arellano JI, Macklis JD, Rakic P. Everything that glitters isn't gold: a critical review of postnatal neural precursor analyses. Cell Stem Cell 2008; 1:612-27. [PMID: 18371403 DOI: 10.1016/j.stem.2007.11.008] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adult neurogenesis research has made enormous strides in the last decade but has been complicated by several failures to replicate promising findings. Prevalent use of highly sensitive methods with inherent sources of error has led to extraordinary conclusions without adequate crossvalidation. Perhaps the biggest culprit is the reliance on molecules involved in DNA synthesis and genetic markers to indicate neuronal neogenesis. In this Protocol Review, we present an overview of common methodological issues in the field and suggest alternative approaches, including viral vectors, siRNA, and inducible transgenic/knockout mice. A multipronged approach will enhance the overall rigor of research on stem cell biology and related fields by allowing increased replication of findings between groups and across systems.
Collapse
Affiliation(s)
- Joshua J Breunig
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
40
|
Skalnikova H, Vodicka P, Gadher SJ, Kovarova H. Proteomics of neural stem cells. Expert Rev Proteomics 2008; 5:175-86. [PMID: 18466050 DOI: 10.1586/14789450.5.2.175] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The isolation of neural stem cells from fetal and adult mammalian CNS and the demonstration of functional neurogenesis in adult CNS have offered perspectives for treatment of many devastating hereditary and acquired neurological diseases. Due to this enormous potential, neural stem cells are a subject of extensive molecular profiling studies with a search for new markers and regulatory pathways governing their self-renewal as opposed to differentiation. Several in-depth proteomic studies have been conducted on primary or immortalized cultures of neural stem cells and neural progenitor cells, and yet more remains to be done. Additionally, neurons and glial cells have been obtained from embryonic stem cells and mesenchymal stem cells, and proteins associated with the differentiation process have been characterized to a certain degree with a view to further investigations. This review summarizes recent findings relevant to the proteomics of neural stem cells and discusses major proteins significantly regulated during neural stem cell differentiation with a view to their future use in cell-based regenerative and reparative therapy.
Collapse
Affiliation(s)
- Helena Skalnikova
- Institute of Animal Physiology & Genetics, Academy of Sciences of the Czech Republic, Rumburska 89, 277 21 Libechov, Czech Republic.
| | | | | | | |
Collapse
|
41
|
Tondreau T, Dejeneffe M, Meuleman N, Stamatopoulos B, Delforge A, Martiat P, Bron D, Lagneaux L. Gene expression pattern of functional neuronal cells derived from human bone marrow mesenchymal stromal cells. BMC Genomics 2008; 9:166. [PMID: 18405367 PMCID: PMC2358905 DOI: 10.1186/1471-2164-9-166] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Accepted: 04/11/2008] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Neuronal tissue has limited potential to self-renew or repair after neurological diseases. Cellular therapies using stem cells are promising approaches for the treatment of neurological diseases. However, the clinical use of embryonic stem cells or foetal tissues is limited by ethical considerations and other scientific problems. Thus, bone marrow mesenchymal stomal cells (BM-MSC) could represent an alternative source of stem cells for cell replacement therapies. Indeed, many studies have demonstrated that MSC can give rise to neuronal cells as well as many tissue-specific cell phenotypes. METHODS BM-MSC were differentiated in neuron-like cells under specific induction (NPBM + cAMP + IBMX + NGF + Insulin). By day ten, differentiated cells presented an expression profile of real neurons. Functionality of these differentiated cells was evaluated by calcium influx through glutamate receptor AMPA3. RESULTS Using microarray analysis, we compared gene expression profile of these different samples, before and after neurogenic differentiation. Among the 1943 genes differentially expressed, genes down-regulated are involved in osteogenesis, chondrogenesis, adipogenesis, myogenesis and extracellular matrix component (tuftelin, AGC1, FADS3, tropomyosin, fibronectin, ECM2, HAPLN1, vimentin). Interestingly, genes implicated in neurogenesis are increased. Most of them are involved in the synaptic transmission and long term potentialisation as cortactin, CASK, SYNCRIP, SYNTL4 and STX1. Other genes are involved in neurite outgrowth, early neuronal cell development, neuropeptide signaling/synthesis and neuronal receptor (FK506, ARHGAP6, CDKRAP2, PMCH, GFPT2, GRIA3, MCT6, BDNF, PENK, amphiregulin, neurofilament 3, Epha4, synaptotagmin). Using real time RT-PCR, we confirmed the expression of selected neuronal genes: NEGR1, GRIA3 (AMPA3), NEF3, PENK and Epha4. Functionality of these neuron-like cells was demonstrated by Ca2+ influx through glutamate receptor channel (AMPA3) in the presence of two agonist glutamate, AMPA or CNQX antagonist. CONCLUSION Our results demonstrate that BM-MSC have the potential to differentiate in neuronal cells with specific gene expression and functional properties. BM-MSC are thus promising candidates for cell-based therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Tatiana Tondreau
- Institut Jules Bordet, Université Libre de Bruxelles, Laboratory of Experimental Hematology, 121, Bd de Waterloo, 1000 Brussels, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Proteomics has evolved, in recent years, into effective tools for basic and applied stem cell research, and has been extensively used to facilitate the identification of changes in signal transduction components, especially with regard to plasticity, proliferation, and differentiation. Several recent reports have also employed proteomic strategies to characterize human mesenchymal stem cells (hMSC) and their differentiated derivatives. Although these approaches have yielded valuable data, the results highlight the fact that only the limited numbers of proteins are characterized at the protein level in these cells, thus necessitating expandable MSC proteome dataset. This review presents, for the first time, an expandable list of MSC proteins, which will function as a starting point for the generation of a comprehensive reference map of their proteome. Also, the better way to bridge current gap between genomics and proteomics study such as integrated proteomic and transcriptomic analyses is discussed.
Collapse
Affiliation(s)
- Hye Won Park
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | | | | |
Collapse
|
43
|
Parolini O, Alviano F, Bagnara GP, Bilic G, Bühring HJ, Evangelista M, Hennerbichler S, Liu B, Magatti M, Mao N, Miki T, Marongiu F, Nakajima H, Nikaido T, Portmann-Lanz CB, Sankar V, Soncini M, Stadler G, Surbek D, Takahashi TA, Redl H, Sakuragawa N, Wolbank S, Zeisberger S, Zisch A, Strom SC. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international Workshop on Placenta Derived Stem Cells. Stem Cells 2007; 26:300-11. [PMID: 17975221 DOI: 10.1634/stemcells.2007-0594] [Citation(s) in RCA: 744] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Placental tissue draws great interest as a source of cells for regenerative medicine because of the phenotypic plasticity of many of the cell types isolated from this tissue. Furthermore, placenta, which is involved in maintaining fetal tolerance, contains cells that display immunomodulatory properties. These two features could prove useful for future cell therapy-based clinical applications. Placental tissue is readily available and easily procured without invasive procedures, and its use does not elicit ethical debate. Numerous reports describing stem cells from different parts of the placenta, using nearly as numerous isolation and characterization procedures, have been published. Considering the complexity of the placenta, an urgent need exists to define, as clearly as possible, the region of origin and methods of isolation of cells derived from this tissue. On March 23-24, 2007, the first international Workshop on Placenta Derived Stem Cells was held in Brescia, Italy. Most of the research published in this area focuses on mesenchymal stromal cells isolated from various parts of the placenta or epithelial cells isolated from amniotic membrane. The aim of this review is to summarize and provide the state of the art of research in this field, addressing aspects such as cell isolation protocols and characteristics of these cells, as well as providing preliminary indications of the possibilities for use of these cells in future clinical applications.
Collapse
Affiliation(s)
- Ornella Parolini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza, Istituto Ospedaliero, Via Bissolati 57, 25124 Brescia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chung N, Jee BK, Chae SW, Jeon YW, Lee KH, Rha HK. HOX gene analysis of endothelial cell differentiation in human bone marrow-derived mesenchymal stem cells. Mol Biol Rep 2007; 36:227-35. [PMID: 17972163 DOI: 10.1007/s11033-007-9171-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2007] [Accepted: 10/19/2007] [Indexed: 12/11/2022]
Abstract
Human bone marrow-derived mesenchymal stem cells (hMSCs) have been shown to possess multilineage differentiation potential. HOX genes function in transcriptional regulators, and are involved in stem cell differentiation. The aim of the present study was to demonstrate HOX genes that are related to angiogenesis. To identify the expression patterns of 37 HOX genes in the endothelial cell differentiation of hMSCs, we analyzed HOX genes through profiling with multiplex RT-PCR. The results showed that the expression patterns of four HOX genes, HOXA7, HOXB3, HOXA3, and HOXB13, significantly changed during angiogenesis. The expression levels of HOXA7 and HOXB3 were dramatically increased, whereas those of HOXA3 and HOXB13 were decreased during endothelial cell differentiation. When further analysis of the expressions of these HOX genes was performed with real-time PCR and an immunoblot assay, the expression patterns were also found to be well-matched with the results of multiplex RT-PCR. Here, we report that HOXA7, HOXB3, HOXA3, and HOXB13 might be involved in the angiogenesis of hMSCs.
Collapse
Affiliation(s)
- Namhyun Chung
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
45
|
Beyer S, Mix E, Hoffrogge R, Lünser K, Völker U, Rolfs A. Neuroproteomics in stem cell differentiation. Proteomics Clin Appl 2007; 1:1513-23. [PMID: 21136647 DOI: 10.1002/prca.200700324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Indexed: 12/31/2022]
Abstract
The term "proteome" is used to describe the entire complement of proteins in a given organism or in a system at a given time. Proteome analysis in neuroscience, also called "neuroproteomics" or "neuromics" is in its initial stage, and shows a deficit of studies in the context of brain development. It is the main objective of this review to illustrate the potential of neuroproteomics as a tool to unravel the differentiation of neural stem or progenitor cells to terminally differentiated neurons. Experimental results regarding the rat striatal progenitor model cell line ST14A are presented to illustrate the large rearrangements of the proteome during the differentiation process of neural progenitor cells and their modification by neurotrophic factors like the glial cell line-derived neurotrophic factor (GDNF). Thereby native stem cells and cells transfected with GDNF gene were investigated at the proliferative state and at seven time points up to 72 h after induction of differentiation. In addition, the immortalized human fetal midbrain stem cell line ReNcell VM was analyzed in order to detect stem cell differentiation associated changes of the protein profile. This review gives also an outlook on technical improvements and perspectives of application of neural stem cell proteomics.
Collapse
Affiliation(s)
- Susanne Beyer
- Neurobiological Laboratory, Department of Neurology, Medical Faculty, University of Rostock, Rostock, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Phinney DG, Prockop DJ. Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair--current views. Stem Cells 2007; 25:2896-902. [PMID: 17901396 DOI: 10.1634/stemcells.2007-0637] [Citation(s) in RCA: 1409] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells or multipotent stromal cells (MSCs) isolated from the bone marrow of adult organisms were initially characterized as plastic adherent, fibroblastoid cells with the capacity to generate heterotopic osseous tissue when transplanted in vivo. In recent years, MSCs or MSC-like cells have been shown to reside within the connective tissue of most organs, and their surface phenotype has been well described. A large number of reports have also indicated that the cells possess the capacity to transdifferentiate into epithelial cells and lineages derived from the neuroectoderm. The broad developmental plasticity of MSCs was originally thought to contribute to their demonstrated efficacy in a wide variety of experimental animal models of disease as well as in human clinical trials. However, new findings suggest that the ability of MSCs to alter the tissue microenvironment via secretion of soluble factors may contribute more significantly than their capacity for transdifferentiation in tissue repair. Herein, we critically evaluate the literature describing the plasticity of MSCs and offer insight into how the molecular and functional heterogeneity of this cell population, which reflects the complexity of marrow stroma as an organ system, may confound interpretation of their transdifferentiation potential. Additionally, we argue that this heterogeneity also provides a basis for the broad therapeutic efficacy of MSCs.
Collapse
Affiliation(s)
- Donald G Phinney
- Center for Gene Therapy, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, Louisiana 70112, USA.
| | | |
Collapse
|
47
|
Fox JM, Chamberlain G, Ashton BA, Middleton J. Recent advances into the understanding of mesenchymal stem cell trafficking. Br J Haematol 2007; 137:491-502. [PMID: 17539772 DOI: 10.1111/j.1365-2141.2007.06610.x] [Citation(s) in RCA: 202] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The use of adult stem cells to regenerate damaged tissue circumvents the moral and technical issues associated with the use of those from an embryonic source. Mesenchymal stem cells (MSC) can be isolated from a variety of tissues, most commonly from the bone marrow, and, although they represent a very small percentage of these cells, are easily expandable. Recently, the use of MSC has provided clinical benefit to patients with osteogenesis imperfecta, graft-versus-host disease and myocardial infarction. The cellular cues that enabled the MSC to be directed to the sites of tissue damage and the mechanisms by which MSC then exert their therapeutic effect are becoming clearer. This review discusses the relative therapeutic importance of the ability of MSC to differentiate into multiple cell lineages or stimulate resident or attracted cells via a paracrine mode of action. It also reviews recent findings that MSC home to damaged tissues in a similar, but somewhat distinct, manner to that of leucocytes via the utilisation of adhesion molecules, such as selectins and integrins, and chemokines and their receptors in a manner reminiscent of leucocytes trafficking from the blood stream to inflammatory sites.
Collapse
Affiliation(s)
- James M Fox
- Arthritis Research Centre, Institute of Science and Technology in Medicine, Keele University at Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK.
| | | | | | | |
Collapse
|
48
|
Abstract
Gene expression analyses of stem cells (SCs) will help to uncover or further define signaling pathways and molecular mechanisms involved in the maintenance of self-renewal, pluripotency, and/or multipotency. In recent years, proteomic approaches have produced a wealth of data identifying proteins and mechanisms involved in SC proliferation and differentiation. Although many proteomics techniques have been developed and improved in peptide and protein separation, as well as mass spectrometry, several important issues, including sample heterogeneity, post-translational modifications, protein-protein interaction, and high-throughput quantification of hydrophobic and low-abundance proteins, still remain to be addressed and require further technical optimization. This review summarizes the methodologies used and the information gathered with proteome analyses of SCs, and it discusses biological and technical challenges for proteomic study of SCs. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
|