1
|
Alexanian AR, Sorokin A, Duersteler M. Dopaminergic progenitors generated by small molecule approach survived, integrated, and promoted functional recovery in (6-OHDA) mouse model of Parkinson's disease. J Neurol Sci 2024; 465:123188. [PMID: 39178824 PMCID: PMC11412743 DOI: 10.1016/j.jns.2024.123188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder resulting from the loss of dopamine-producing neurons in the brain, causing motor symptoms like tremors and stiffness. Although current treatments like medication and deep brain stimulation can alleviate symptoms, they don't address the root cause of neuron loss. Therefore, cell replacement therapy emerges as a promising treatment strategy. However, the generation of engraftable dopaminergic (DA) cells in clinically relevant quantities is still a challenge. Recent advances in cell reprogramming technologies open up vast possibilities to produce patient-specific cells of a desired type in therapeutic quantities. The main cell reprogramming strategies involve the enforced expression of individual or sets of genes through viral transduction or transfection, or through small molecules, known as the chemical approach, which is a much easier and safer method. In our previous studies, using a small molecule approach (combinations of epigenetic modifiers and SMAD inhibitors such asDorsomorphin and SB431542), we have been able to generate DA progenitors from human mesenchymal stem cells (hMSCs). The aim of this study was to further improve the method for the generation of DA progenitors and to test their therapeutic effect in an animal model of Parkinson's. The results showed that the addition of an autophagy enhancer (AE) to our DA cell induction protocol further increased the yield of DA progenitor cells. The results also showed that DA progenitors transplanted into the mouse model of PD survived, integrated, and improved PD motor symptoms. These data suggest that chemically-produced DA cells can be very promising and safe cellular therapeutics for PD.
Collapse
Affiliation(s)
- Arshak R Alexanian
- Cell Reprogramming & Therapeutics LLC, Wauwatosa (Milwaukee County), WI 53226, USA; Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, United States of America.
| | - Andrey Sorokin
- Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, United States of America
| | - Megan Duersteler
- Cell Reprogramming & Therapeutics LLC, Wauwatosa (Milwaukee County), WI 53226, USA
| |
Collapse
|
2
|
Alexanian AR. Combination of the modulators of epigenetic machinery and specific cell signaling pathways as a promising approach for cell reprogramming. Mol Cell Biochem 2022; 477:2309-2317. [PMID: 35503191 DOI: 10.1007/s11010-022-04442-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/08/2022] [Indexed: 11/27/2022]
Abstract
During embryogenesis and further development, mammalian epigenome undergoes global remodeling, which leads to the emergence of multiple fate-restricted cell lines as well as to their further differentiation into different specialized cell types. There are multiple lines of evidence suggesting that all these processes are mainly controlled by epigenetic mechanisms such as DNA methylation, histone covalent modifications, and the regulation of ATP-dependent remolding of chromatin structure. Based on the histone code hypothesis, distinct chromatin covalent modifications can lead to functionally distinct chromatin structures and thus distinctive gene expression that determine the fate of the cells. A large amount of recently accumulated data showed that small molecule biologically active compounds that involved in the regulation of chromatin structure and function in discriminative signaling environments can promote changes in cells fate. These data suggest that agents that involved in the regulation of chromatin modifying enzymes combined with factors that modulate specific cell signaling pathways could be effective tools for cell reprogramming. The goal of this review is to gather the most relevant and most recent literature that supports this proposition.
Collapse
Affiliation(s)
- Arshak R Alexanian
- Cell Reprogramming & Therapeutics LLC, 10437 Innovation drive, Suite 321, Wauwatosa, WI, 53226, USA.
| |
Collapse
|
3
|
Wang N, Ji X, Wu Y, Zhou S, Peng H, Wang J, Yu S, Zhang J. The Different Molecular Code in Generation of Dopaminergic Neurons from Astrocytes and Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms222212141. [PMID: 34830023 PMCID: PMC8622032 DOI: 10.3390/ijms222212141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 11/26/2022] Open
Abstract
Transplantation of exogenous dopaminergic (DA) neurons is an alternative strategy to replenish DA neurons that have lost along the course of Parkinson’s disease (PD). From the perspective of ethical acceptation, the source limitations, and the intrinsic features of PD pathology, astrocytes (AS) and mesenchymal stem cells (MSCs) are the two promising candidates of DA induction. In the present study, we induced AS or MSCs primary culture by the combination of the classical transcription-factor cocktails Mash1, Lmx1a, and Nurr1 (MLN), the chemical cocktails (S/C/D), and the morphogens SHH, FGF8, and FGF2 (S/F8/F2); the efficiency of induction into DA neurons was further analyzed by using immunostaining against the DA neuronal markers. AS could be efficiently converted into the DA neurons in vitro by the transcriptional regulation of MLN, and the combination with S/C/D or S/F8/F2 further increased the conversion efficiency. In contrast, MSCs from umbilical cord (UC-MSCs) or adipose tissue (AD-MSCs) showed moderate TH immunoreactivity after the induction with S/F8/F2 instead of with MLN or S/C/D. Our data demonstrated that AS and MSCs held lineage-specific molecular codes on the induction into DA neurons and highlighted the unique superiority of AS in the potential of cell replacement therapy for PD.
Collapse
Affiliation(s)
- Nana Wang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
| | - Xingrui Ji
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
| | - Yue Wu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
- Zhengzhou Zhongke Institute of Biomedical Engineering and Technology, Zhengzhou 450001, China
| | - Shaocong Zhou
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
| | - Huiyu Peng
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
| | - Jingwen Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
- Zhengzhou Zhongke Institute of Biomedical Engineering and Technology, Zhengzhou 450001, China
| | - Shuang Yu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
- Correspondence: (S.Y.); (J.Z.); Tel.: +86-13951103916 (S.Y.); +86-13913161723 (J.Z.)
| | - Jingzhong Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
- Zhengzhou Zhongke Institute of Biomedical Engineering and Technology, Zhengzhou 450001, China
- Correspondence: (S.Y.); (J.Z.); Tel.: +86-13951103916 (S.Y.); +86-13913161723 (J.Z.)
| |
Collapse
|
4
|
Liu D, Bobrovskaya L, Zhou XF. Cell Therapy for Neurological Disorders: The Perspective of Promising Cells. BIOLOGY 2021; 10:1142. [PMID: 34827135 PMCID: PMC8614777 DOI: 10.3390/biology10111142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022]
Abstract
Neurological disorders are big public health challenges that are afflicting hundreds of millions of people around the world. Although many conventional pharmacological therapies have been tested in patients, their therapeutic efficacies to alleviate their symptoms and slow down the course of the diseases are usually limited. Cell therapy has attracted the interest of many researchers in the last several decades and has brought new hope for treating neurological disorders. Moreover, numerous studies have shown promising results. However, none of the studies has led to a promising therapy for patients with neurological disorders, despite the ongoing and completed clinical trials. There are many factors that may affect the outcome of cell therapy for neurological disorders due to the complexity of the nervous system, especially cell types for transplantation and the specific disease for treatment. This paper provides a review of the various cell types from humans that may be clinically used for neurological disorders, based on their characteristics and current progress in related studies.
Collapse
Affiliation(s)
| | | | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; (D.L.); (L.B.)
| |
Collapse
|
5
|
Salehi-Pourmehr H, Hajebrahimi S, Rahbarghazi R, Pashazadeh F, Mahmoudi J, Maasoumi N, Sadigh-Eteghad S. Stem Cell Therapy for Neurogenic Bladder Dysfunction in Rodent Models: A Systematic Review. Int Neurourol J 2020; 24:241-257. [PMID: 33017895 PMCID: PMC7538284 DOI: 10.5213/inj.2040058.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/23/2020] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Neurogenic bladder dysfunction (NGB) has an impact on the quality of life, which made it an important research subject in preclinical studies. The present review investigates the effect of stem cell (SC) therapy on bladder functional recovery after the onset of spinal cord injury (SCI), multiple sclerosis (MS), Parkinson disease (PD), and stroke in rodent models. METHODS All experiments evaluated the regenerative potential of SC on the management of NGB in rodent models up to June 2019, were included. From 1,189 relevant publications, 20 studies met our inclusion criteria of which 15 were conducted on SCI, 2 on PD, 2 on stroke, and 1 on MS in the rodent models. We conducted a meta-analysis on SCI experiments and for other neurological diseases, detailed urodynamic findings were reported. RESULTS The common SC sources used for therapeutical purposes were neural progenitor cells, bone marrow mesenchymal SCs, human amniotic fluid SCs, and human umbilical cord blood SCs. There was a significant improvement of micturition pressure in both contusion and transaction SCI models 4 and 8 weeks post-SC transplantation. Residual urine volume, micturition volume, and bladder capacity were improved 28 days after SC transplantation only in the transaction model of SCI. Nonvoiding contraction recovered only in 56 days post-cell transplantation in the contusion model. CONCLUSION Partial bladder recovery has been evident after SC therapy in SCI models. Due to limitations in the number of studies in other neurological diseases, additional studies are necessary to confirm the detailed mechanism for bladder recovery.
Collapse
Affiliation(s)
- Hanieh Salehi-Pourmehr
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute (JBI) Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sakineh Hajebrahimi
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute (JBI) Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
- Urology Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Pashazadeh
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute (JBI) Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narjes Maasoumi
- University Hospital Southampton, Southampton, United Kingdom
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- East-Azerbaijan Comprehensive Stroke Program, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Persian Medicine, Faculty of Persian Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Kruminis-Kaszkiel E, Osowski A, Bejer-Oleńska E, Dziekoński M, Wojtkiewicz J. Differentiation of Human Mesenchymal Stem Cells from Wharton's Jelly Towards Neural Stem Cells Using A Feasible and Repeatable Protocol. Cells 2020; 9:cells9030739. [PMID: 32192154 PMCID: PMC7140706 DOI: 10.3390/cells9030739] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022] Open
Abstract
The transplantation of neural stem cells (NSCs) capable of regenerating to the cells of the central nervous system (CNS) is a promising strategy in the treatment of CNS diseases and injury. As previous studies have highlighted mesenchymal stem cells (MSCs) as a source of NSCs, this study aimed to develop a feasible, efficient, and reproducible method for the neural induction of MSCs isolated from Wharton's jelly (hWJ-MSCs). We induced neural differentiation in a monolayer culture using epidermal growth factor, basic fibroblast growth factor, N2, and B27 supplements. This resulted in a homogenous population of proliferating cells that expressed certain neural markers at both the protein and mRNA levels. Flow cytometry and immunocytochemistry confirmed the expression of neural markers: nestin, sex-determining region Y (SRY) box 1 and 2 (SOX1 and SOX2), microtubule-associated protein 2 (MAP2), and glial fibrillary acidic protein (GFAP). The qRT-PCR analysis revealed significantly enhanced expression of nestin and MAP2 in differentiated cells. This study confirms that it is possible to generate NSCs-like cells from hWJ-MSCs in a 2D culture using a practical method. However, the therapeutic effectiveness of such differentiated cells should be extended to confirm the terminal differentiation ability and electrophysiological properties of neurons derived from them.
Collapse
Affiliation(s)
- Ewa Kruminis-Kaszkiel
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
- Correspondence:
| | - Adam Osowski
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| | - Ewa Bejer-Oleńska
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| | - Mariusz Dziekoński
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| |
Collapse
|
7
|
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
|
8
|
Dahbour S, Jamali F, Alhattab D, Al-Radaideh A, Ababneh O, Al-Ryalat N, Al-Bdour M, Hourani B, Msallam M, Rasheed M, Huneiti A, Bahou Y, Tarawneh E, Awidi A. Mesenchymal stem cells and conditioned media in the treatment of multiple sclerosis patients: Clinical, ophthalmological and radiological assessments of safety and efficacy. CNS Neurosci Ther 2017; 23:866-874. [PMID: 28961381 PMCID: PMC5698713 DOI: 10.1111/cns.12759] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 08/16/2017] [Accepted: 09/01/2017] [Indexed: 12/30/2022] Open
Abstract
AIMS This open-label prospective phase I/IIa clinical study used autologous bone marrow-derived mesenchymal stromal cells (BM-MSCs) followed by mesenchymal stromal cells conditioned media (MSC-CM) for the first time to treat multiple sclerosis (MS) patients. The primary goal was to assess the safety and feasibility and the secondary was efficacy. The correlation between the MSC-CM content and treatment outcome was investigated. METHODS Ten MS patients who failed conventional therapy were enrolled. Adverse events were recorded to assess safety. The Expanded Disability Status Scale (EDSS) was the primary efficacy measurement, the secondary included clinical (25WFT, 9-PHT), cognitive (MMS), ophthalmology (OCT, VEP), and radiological (MRI lesion and volume) tests. The MSCs-CM concentration of 27 inflammatory biomarkers was investigated. RESULTS The treatment protocol was well tolerated by patients. There was an overall trend of improvement in all the tests, except the lesion volume which increased significantly. A decrease of 4 and 3.5 points on the EDSS was achieved in two patients. We report a correlation between a decreased lesion number at baseline and higher IL-6, IL-8, and VEGF MSC-CM content. CONCLUSION The used protocol was safe and feasible with possible efficacy. The addition of MSC-CM could be related to the magnitude of EDSS improvement observed.
Collapse
Affiliation(s)
- Said Dahbour
- Department of Neurology, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Fatima Jamali
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Dana Alhattab
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Ali Al-Radaideh
- Department of Medical Imaging, Faculty of Allied Health Sciences, Hashemite University, Zarqa, Jordan
| | - Osama Ababneh
- Department of Ophthalmology, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Nosaiba Al-Ryalat
- Department of Diagnostic Radiology and Nuclear Medicine, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Muawyeh Al-Bdour
- Department of Ophthalmology, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Bayan Hourani
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Mohammed Msallam
- Department of Ophthalmology, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Murad Rasheed
- Department of Neurology, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Ammar Huneiti
- Department of Computer Information Systems, King Abdullah II School for Information Technology, The University of Jordan, Jordan
| | - Yacoub Bahou
- Department of Neurology, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Emad Tarawneh
- Department of Diagnostic Radiology and Nuclear Medicine, Jordan University Hospital, The University of Jordan, Amman, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan.,Department of Medicine and Hematology, Jordan University Hospital, University of Jordan, Amman, Jordan
| |
Collapse
|
9
|
Li Q, Zhang S, Zheng Y, Wen H, Han X, Zhang M, Guan W. Differentiation potential of neural stem cells derived from fetal sheep. Anim Cells Syst (Seoul) 2017; 21:233-240. [PMID: 30460074 PMCID: PMC6138347 DOI: 10.1080/19768354.2017.1354915] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 06/14/2017] [Accepted: 07/10/2017] [Indexed: 11/30/2022] Open
Abstract
Neural stem cells (NSCs) are multipotent stem cells that can differentiate into many cell types in vitro. In this study, we isolated and established an NSC line from fetal Ovis aries. Based on the results of immunofluorescence staining, NSCs expressed Nestin, Pax6 and MAP2. Moreover, a reverse transcription–polymerase chain reaction assay was used to biologically characterize the cell line. NSCs were induced to differentiate into neurogenic cells in vitro. They expressed MAP2, glial fibrillary acidic protein (GFAP) and myelin basic protein (MBP). In this study, we successfully isolated and cultivated NSCs from the hippocampal tissue of fetal sheep. NSCs not only displayed a self-renewal capacity but also had the potential to differentiate into neurons and glial cells. This study provided valuable experimental data for NSC transplant research.
Collapse
Affiliation(s)
- Qian Li
- College of Wildlife Resources, Northeast Forestry University, Harbin, PR China
| | - Shuang Zhang
- Scientific Experiment Research Center, Harbin Institute of Physical Education, Harbin, PR China
| | - Yanjie Zheng
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Hebao Wen
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Xiao Han
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Minghai Zhang
- College of Wildlife Resources, Northeast Forestry University, Harbin, PR China
| | - Weijun Guan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, PR China
| |
Collapse
|
10
|
Alexanian AR. Epigenetic modulators promote mesenchymal stem cell phenotype switches. Int J Biochem Cell Biol 2015; 64:190-4. [PMID: 25936755 DOI: 10.1016/j.biocel.2015.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/14/2015] [Accepted: 04/21/2015] [Indexed: 01/18/2023]
Abstract
Discoveries in recent years have suggested that some tissue specific adult stem cells in mammals might have the ability to differentiate into cell types from different germ layers. This phenomenon has been referred to as stem cell transdifferentiation or plasticity. Despite controversy, the current consensus holds that transdifferentiation does occur in mammals, but only within a limited range. Understanding the mechanisms that underlie the switches in phenotype and development of the methods that will promote such type of conversions can open up endless possibilities for regenerative medicine. Epigenetic control contributes to various processes that lead to cellular plasticity and DNA and histone covalent modifications play a key role in these processes. Recently, we have been able to convert human mesenchymal stem cells (hMSCs) into neural-like cells by exposing cells to epigenetic modifiers and neural inducing factors. The goal of this study was to investigate the stability and plasticity of these transdifferentiated cells. To this end, neurally induced MSCs (NI-hMSCs) were exposed to adipocyte inducing factors. Grown for 24-48 h in fat induction media NI-hMSCs reversed their morphology into fibroblast-like cells and regained their proliferative properties. After 3 weeks approximately 6% of hMSCs differentiated into multilocular or plurivacuolar adipocyte cells that demonstrated by Oil Red O staining. Re-exposure of these cultures or the purified adipocytes to neural induction medium induced the cells to re-differentiate into neuronal-like cells. These data suggest that cell plasticity can be manipulated by the combination of small molecule modulators of chromatin modifying enzymes and specific cell signaling pathways.
Collapse
Affiliation(s)
- Arshak R Alexanian
- Cell Reprogramming & Therapeutics LLC, W229 N1870 Westwood Drive, Waukesha, WI 53186 United States.
| |
Collapse
|
11
|
Zhang R, Zhang K, Li J, Liu Q, Xie J. In vivo tracking of neuronal-like cells by magnetic resonance in rabbit models of spinal cord injury. Neural Regen Res 2014; 8:3373-81. [PMID: 25206659 PMCID: PMC4146005 DOI: 10.3969/j.issn.1673-5374.2013.36.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/17/2013] [Indexed: 01/25/2023] Open
Abstract
In vitro experiments have demonstrated that neuronal-like cells derived from bone marrow mesenchymal stem cells can survive, migrate, integrate and help to restore the function and behaviors of spinal cord injury models, and that they may serve as a suitable approach to treating spinal cord injury. However, it is very difficult to track transplanted cells in vivo. In this study, we injected superparamagnetic iron oxide-labeled neuronal-like cells into the subarachnoid space in a rabbit model of spinal cord injury. At 7 days after cell transplantation, a small number of dot-shaped low signal intensity shadows were observed in the spinal cord injury region, and at 14 days, the number of these shadows increased on T2-weighted imaging. Perl's Prussian blue staining detected dot-shaped low signal intensity shadows in the spinal cord injury region, indicative of superparamagnetic iron oxide nanoparticle-labeled cells. These findings suggest that transplanted neuronal-like cells derived from bone marrow mesenchymal stem cells can migrate to the spinal cord injury region and can be tracked by magnetic resonance in vivo. Magnetic resonance imaging represents an efficient noninvasive technique for visually tracking transplanted cells in vivo.
Collapse
Affiliation(s)
- Ruiping Zhang
- Department of Radiology, First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Kun Zhang
- Department of Radiology, Medical Imaging Centre, Navy General Hospital, Beijing 100048, China
| | - Jianding Li
- Department of Radiology, First Hospital, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Qiang Liu
- Department of Orthopedics, Shanxi Dayi Hospital, Taiyuan 030032, Shanxi Province, China
| | - Jun Xie
- Department of Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| |
Collapse
|
12
|
Yang C, Dai W, Chen H, Wu B. Application of human bone marrow-derived mesenchymal stem cells in the treatment of radiation-induced Gastrointestinal syndrome. SCIENCE CHINA-LIFE SCIENCES 2014; 57:1177-82. [PMID: 25205377 DOI: 10.1007/s11427-014-4721-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 10/22/2013] [Indexed: 01/14/2023]
Abstract
Nuclear accidents and terrorism present a serious threat for mass casualty. Accidental or intended radiation exposure leads to radiation-induced gastrointestinal (GI) syndrome. However, currently there are no approved medical countermeasures for GI syndrome. Thus, developing novel treatments for GI syndrome is urgent. Mesenchymal stem cells (MSCs) derived from bone marrow are a subset of multipotent adult somatic stem cells that have the ability to undergo self-renewal, proliferation and pluripotent differentiation. MSCs have advantages over other stem cells; they can be easily isolated from patients or donors, readily expanded ex vivo, and they possess reparative and immunomodulatory properties. Moreover, MSCs have been shown to be powerful tools in gene therapy and can be effectively transduced with vectors containing therapeutic genes. Therefore, the therapeutic potential of MSCs has been brought into the spotlight for the clinical treatment of GI syndrome. In this review, we discuss the possible role of MSCs in radiation-induced GI syndrome.
Collapse
Affiliation(s)
- Chao Yang
- Gastrointestinal Department of Southern Building, General Hospital of Chinese PLA, Beijing, 100853, China
| | | | | | | |
Collapse
|
13
|
Mesenchymal stem cells, neural lineage potential, heparan sulfate proteoglycans and the matrix. Dev Biol 2014; 388:1-10. [DOI: 10.1016/j.ydbio.2014.01.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 01/08/2014] [Accepted: 01/30/2014] [Indexed: 12/23/2022]
|
14
|
Hogberg HT, Bressler J, Christian KM, Harris G, Makri G, O'Driscoll C, Pamies D, Smirnova L, Wen Z, Hartung T. Toward a 3D model of human brain development for studying gene/environment interactions. Stem Cell Res Ther 2013; 4 Suppl 1:S4. [PMID: 24564953 PMCID: PMC4029162 DOI: 10.1186/scrt365] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This project aims to establish and characterize an in vitro model of the developing human brain for the purpose of testing drugs and chemicals. To accurately assess risk, a model needs to recapitulate the complex interactions between different types of glial cells and neurons in a three-dimensional platform. Moreover, human cells are preferred over cells from rodents to eliminate cross-species differences in sensitivity to chemicals. Previously, we established conditions to culture rat primary cells as three-dimensional aggregates, which will be humanized and evaluated here with induced pluripotent stem cells (iPSCs). The use of iPSCs allows us to address gene/environment interactions as well as the potential of chemicals to interfere with epigenetic mechanisms. Additionally, iPSCs afford us the opportunity to study the effect of chemicals during very early stages of brain development. It is well recognized that assays for testing toxicity in the developing brain must consider differences in sensitivity and susceptibility that arise depending on the time of exposure. This model will reflect critical developmental processes such as proliferation, differentiation, lineage specification, migration, axonal growth, dendritic arborization and synaptogenesis, which will probably display differences in sensitivity to different types of chemicals. Functional endpoints will evaluate the complex cell-to-cell interactions that are affected in neurodevelopment through chemical perturbation, and the efficacy of drug intervention to prevent or reverse phenotypes. The model described is designed to assess developmental neurotoxicity effects on unique processes occurring during human brain development by leveraging human iPSCs from diverse genetic backgrounds, which can be differentiated into different cell types of the central nervous system. Our goal is to demonstrate the feasibility of the personalized model using iPSCs derived from individuals with neurodevelopmental disorders caused by known mutations and chromosomal aberrations. Notably, such a human brain model will be a versatile tool for more complex testing platforms and strategies as well as research into central nervous system physiology and pathology.
Collapse
Affiliation(s)
- Helena T Hogberg
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| | - Joseph Bressler
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
- Hugo Moser Institute at the Kennedy Krieger, Johns Hopkins University, Bloomberg School of Public Health, 707 N. Broadway, Baltimore, MD 21205, USA
| | - Kimberly M Christian
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University, School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Georgina Harris
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| | - Georgia Makri
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University, School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Cliona O'Driscoll
- Hugo Moser Institute at the Kennedy Krieger, Johns Hopkins University, Bloomberg School of Public Health, 707 N. Broadway, Baltimore, MD 21205, USA
| | - David Pamies
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| | - Lena Smirnova
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| | - Zhexing Wen
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University, School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| |
Collapse
|
15
|
Funk RT, Alexanian AR. Enhanced dopamine release by mesenchymal stem cells reprogrammed neuronally by the modulators of SMAD signaling, chromatin modifying enzymes, and cyclic adenosine monophosphate levels. Transl Res 2013; 162:317-23. [PMID: 24001408 DOI: 10.1016/j.trsl.2013.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/07/2013] [Accepted: 08/11/2013] [Indexed: 01/28/2023]
Abstract
Recently, using the chemical genetics approach for cell reprogramming, via the combination of small molecule modulators of chromatin modifying enzymes, specific SMAD signaling pathways, and cyclic adenosine monophosphate levels, we have been able to generate neuronallike cells predominantly positive to mature neuronal and dopaminergic markers. This study aimed to characterize further the dopaminergic properties of neurally induced (NI) human bone marrow-derived mesenchymal stem cells (hMSCs) and to determine whether addition of sonic hedgehog (SHH)/fibroblast growth factor 8 (FGF8) to NI medium could promote further dopaminergic maturation. Dopaminergic differentiation was evaluated by immunocytochemistry, reverse transcription-polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay. Results demonstrated that release of dopamine by NI-hMSCs differentiated with SMAD inhibitor supplementation significantly increased from picogram to nanogram levels, with a tendency of further increase when supplemented by SHH/FGF8. Direct generation of dopaminergic cells from adult hMSCs by using this reprogramming approach may have significant implications for understanding the mechanism underlying cell plasticity and may open new potentialities for cell replacement therapies.
Collapse
Affiliation(s)
- Ryan T Funk
- Neuroscience Research Laboratories, Department of Neurosurgery, VA Medical Center, Milwaukee, Wisc
| | | |
Collapse
|
16
|
Bai WF, Xu WC, Feng Y, Huang H, Li XP, Deng CY, Zhang MS. Fifty-Hertz electromagnetic fields facilitate the induction of rat bone mesenchymal stromal cells to differentiate into functional neurons. Cytotherapy 2013; 15:961-70. [DOI: 10.1016/j.jcyt.2013.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 03/08/2013] [Indexed: 12/21/2022]
|
17
|
Alexanian AR, Liu QS, Zhang Z. Enhancing the efficiency of direct reprogramming of human mesenchymal stem cells into mature neuronal-like cells with the combination of small molecule modulators of chromatin modifying enzymes, SMAD signaling and cyclic adenosine monophosphate levels. Int J Biochem Cell Biol 2013; 45:1633-8. [PMID: 23665234 DOI: 10.1016/j.biocel.2013.04.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/18/2013] [Accepted: 04/25/2013] [Indexed: 01/07/2023]
Abstract
Advances in cell reprogramming technologies to generate patient-specific cells of a desired type will revolutionize the field of regenerative medicine. While several cell reprogramming methods have been developed over the last decades, the majority of these technologies require the exposure of cell nuclei to reprogramming large molecules via transfection, transduction, cell fusion or nuclear transfer. This raises several technical, safety and ethical issues. Chemical genetics is an alternative approach for cell reprogramming that uses small, cell membrane penetrable substances to regulate multiple cellular processes including cell plasticity. Recently, using the combination of small molecules that are involved in the regulation chromatin structure and function and agents that favor neural differentiation we have been able to generate neural-like cells from human mesenchymal stem cells. In this study, to improve the efficiency of neuronal differentiation and maturation, two specific inhibitors of SMAD signaling (SMAD1/3 and SMAD3/5/8) that play an important role in neuronal differentiation of embryonic stem cells, were added to our previous neural induction recipe. Results demonstrated that human mesenchymal stem cells grown in this culture conditions exhibited higher expression of several mature neuronal genes, formed synapse-like structures and exerted electrophysiological properties of differentiating neural stem cells. Thus, an efficient method for production of mature neuronal-like cells from human adult bone marrow derived mesenchymal stem cells has been developed. We concluded that specific combinations of small molecules that target specific cell signaling pathways and chromatin modifying enzymes could be a promising approach for manipulation of adult stem cell plasticity.
Collapse
Affiliation(s)
- Arshak R Alexanian
- Neuroscience Research Laboratories, Department of Neurosurgery, VA Medical Center-Research 151, Milwaukee, WI 53295, United States.
| | | | | |
Collapse
|
18
|
Falavigna A, da Costa JC. Mesenchymal autologous stem cells. World Neurosurg 2013; 83:236-50. [PMID: 23402865 DOI: 10.1016/j.wneu.2013.02.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 01/24/2013] [Accepted: 02/05/2013] [Indexed: 02/07/2023]
Abstract
The use of cell-based therapies for spinal cord injuries has recently gained prominence as a potential therapy or component of a combination strategy. Experimental and clinical studies have been performed using mesenchymal stem cell therapy to treat spinal cord injuries with encouraging results. However, there have been reports on the adverse effects of these stem cell-based therapies, especially in the context of tumor modulation. This article surveys the literature relevant to the potential of mesenchymal autologous stem cells for spinal cord injuries and their clinical implications.
Collapse
Affiliation(s)
- Asdrubal Falavigna
- Department of Neurosurgery, Medical School of the University of Caxias do Sul, Caxias do Sul, Brazil.
| | - Jaderson Costa da Costa
- Neurology Service and Instituto do Cérebro, Pontifical Catholic University of Rio Grande do Sul, Brazil
| |
Collapse
|
19
|
Zhang Z, Alexanian AR. Dopaminergic-like cells from epigenetically reprogrammed mesenchymal stem cells. J Cell Mol Med 2012; 16:2708-14. [PMID: 22681532 PMCID: PMC4118239 DOI: 10.1111/j.1582-4934.2012.01591.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/19/2012] [Indexed: 01/09/2023] Open
Abstract
A number of recent studies have examined the ability of stem cells derived from different sources to differentiate into dopamine-producing cells and ameliorate behavioural deficits in Parkinsonian models. Recently, using the approach of cell reprogramming by small cell-permeable biological active compounds that involved in the regulation of chromatin structure and function, and interfere with specific cell signalling pathways that promote neural differentiation we have been able to generate neural-like cells from human bone marrow (BM)-derived MSCs (hMSCs). Neurally induced hMSCs (NI-hMSCs) exhibited several neural properties and exerted beneficial therapeutic effect on tissue preservation and locomotor recovery in spinal cord injured rats. In this study, we aimed to determine whether hMSCs neuralized by this approach can generate dopaminergic (DA) neurons. Immunocytochemisty studies showed that approximately 50-60% of NI-hMSCs expressed early and late dopaminergic marker such as Nurr-1 and TH that was confirmed by Western blot. ELISA studies showed that NI-hMSCs also secreted neurotrophins and dopamine. Hypoxia preconditioning prior to neural induction increased hMSCs proliferation, viability, expression TH and the secretion level of dopamine induced by ATP. Taken together, these studies demonstrated that hMSCs neurally modified by this original approach can be differentiated towards DA-like neurons.
Collapse
Affiliation(s)
- Zhiying Zhang
- Department of Neurosurgery, Neuroscience Research Labs, Medical College of Wisconsin, VAMCMilwaukee, WI, USA
| | - Arshak R Alexanian
- Department of Neurosurgery, Neuroscience Research Labs, Medical College of Wisconsin, VAMCMilwaukee, WI, USA
| |
Collapse
|
20
|
Zhang Z, Alexanian AR. The neural plasticity of early-passage human bone marrow-derived mesenchymal stem cells and their modulation with chromatin-modifying agents. J Tissue Eng Regen Med 2012; 8:407-13. [DOI: 10.1002/term.1535] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 02/17/2012] [Accepted: 04/10/2012] [Indexed: 01/18/2023]
Affiliation(s)
- Zhiying Zhang
- Neuroscience Research Laboratories, Department of Neurosurgery; VA Medical Center; Milwaukee WI USA
| | - Arshak R. Alexanian
- Neuroscience Research Laboratories, Department of Neurosurgery; VA Medical Center; Milwaukee WI USA
| |
Collapse
|
21
|
Prasongchean W, Ferretti P. Autologous stem cells for personalised medicine. N Biotechnol 2012; 29:641-50. [PMID: 22561284 DOI: 10.1016/j.nbt.2012.04.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 04/17/2012] [Accepted: 04/19/2012] [Indexed: 01/11/2023]
Abstract
Increasing understanding of stem cell biology, the ability to reprogramme differentiated cells to a pluripotent state and evidence of multipotency in certain adult somatic stem cells has opened the door to exciting therapeutic advances as well as a great deal of regulatory and ethical issues. Benefits will come from the possibility of modelling human diseases and develop individualised therapies, and from their use in transplantation and bioengineering. The use of autologous stem cells is highly desirable, as it avoids the problem of tissue rejection, and also reduces ethical and regulatory issues. Identification of the most appropriate cell sources for different potential applications, development of appropriate clinical grade methodologies and large scale well controlled clinical trials will be essential to assess safety and value of cell based therapies, which have been generating much hope, but are by and large not yet close to becoming standard clinical practice. We briefly discuss stem cells in the context of tissue repair and regenerative medicine, with a focus on individualised clinical approaches, and give examples of sources of autologous cells with potential for clinical intervention.
Collapse
|
22
|
Liu Y, Jiang X, Zhang X, Chen R, Sun T, Fok KL, Dong J, Tsang LL, Yi S, Ruan Y, Guo J, Yu MK, Tian Y, Chung YW, Yang M, Xu W, Chung CM, Li T, Chan HC. Dedifferentiation-Reprogrammed Mesenchymal Stem Cells with Improved Therapeutic Potential. Stem Cells 2011; 29:2077-2089. [DOI: 10.1002/stem.764] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AbstractStem cell transplantation has been shown to improve functional outcome in degenerative and ischemic disorders. However, low in vivo survival and differentiation potential of the transplanted cells limits their overall effectiveness and thus clinical usage. Here we show that, after in vitro induction of neuronal differentiation and dedifferentiation, on withdrawal of extrinsic factors, mesenchymal stem cells (MSCs) derived from bone marrow, which have already committed to neuronal lineage, revert to a primitive cell population (dedifferentiated MSCs) retaining stem cell characteristics but exhibiting a reprogrammed phenotype distinct from their original counterparts. Of therapeutic interest, the dedifferentiated MSCs exhibited enhanced cell survival and higher efficacy in neuronal differentiation compared to unmanipulated MSCs both in vitro and in vivo, with significantly improved cognition function in a neonatal hypoxic–ischemic brain damage rat model. Increased expression of bcl-2 family proteins and microRNA-34a appears to be the important mechanism giving rise to this previously undefined stem cell population that may provide a novel treatment strategy with improved therapeutic efficacy.
Collapse
Affiliation(s)
- Yang Liu
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
- Children's Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xiaohua Jiang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University-the Chinese University of Hong Kong, Guangzhou, People's Republic of China
| | - Xiaohu Zhang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Rui Chen
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tingting Sun
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kin Lam Fok
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jianda Dong
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lai Ling Tsang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Shaoqiong Yi
- Department of Applied Molecular Biology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Yechun Ruan
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jinghui Guo
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Mei Kuen Yu
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuemin Tian
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yiu Wa Chung
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Mo Yang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Wenming Xu
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
- Sichuan University–the Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Women's and Children's Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chin Man Chung
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tingyu Li
- Children's Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Hsiao Chang Chan
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University-the Chinese University of Hong Kong, Guangzhou, People's Republic of China
| |
Collapse
|
23
|
Wilkinson AE, McCormick AM, Leipzig ND. Central Nervous System Tissue Engineering: Current Considerations and Strategies. ACTA ACUST UNITED AC 2011. [DOI: 10.2200/s00390ed1v01y201111tis008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
24
|
Zhang Z, Maiman DJ, Kurpad SN, Crowe MJ, Alexanian AR. Feline Bone Marrow-Derived Mesenchymal Stem Cells Express Several Pluripotent and Neural Markers and Easily Turn into Neural-Like Cells by Manipulation with Chromatin Modifying Agents and Neural Inducing Factors. Cell Reprogram 2011; 13:385-90. [DOI: 10.1089/cell.2011.0007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Zhiying Zhang
- Department of Neurosurgery, Neuroscience Research Laboratory, VA Medical Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Dennis J. Maiman
- Department of Neurosurgery, Neuroscience Research Laboratory, VA Medical Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Shekar N. Kurpad
- Department of Neurosurgery, Neuroscience Research Laboratory, VA Medical Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Maria J. Crowe
- Department of Neurosurgery, Neuroscience Research Laboratory, VA Medical Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Arshak R. Alexanian
- Department of Neurosurgery, Neuroscience Research Laboratory, VA Medical Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
25
|
Petschnik AE, Fell B, Tiede S, Habermann JK, Pries R, Kruse C, Danner S. A novel xenogeneic co-culture system to examine neuronal differentiation capability of various adult human stem cells. PLoS One 2011; 6:e24944. [PMID: 21935488 PMCID: PMC3173484 DOI: 10.1371/journal.pone.0024944] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 08/24/2011] [Indexed: 12/21/2022] Open
Abstract
Background Targeted differentiation of stem cells is mainly achieved by the sequential administration of defined growth factors and cytokines, although these approaches are quite artificial, cost-intensive and time-consuming. We now present a simple xenogeneic rat brain co-culture system which supports neuronal differentiation of adult human stem cells under more in vivo-like conditions. Methods and Findings This system was applied to well-characterized stem cell populations isolated from human skin, parotid gland and pancreas. In addition to general multi-lineage differentiation potential, these cells tend to differentiate spontaneously into neuronal cell types in vitro and are thus ideal candidates for the introduced co-culture system. Consequently, after two days of co-culture up to 12% of the cells showed neuronal morphology and expressed corresponding markers on the mRNA and protein level. Additionally, growth factors with the ability to induce neuronal differentiation in stem cells could be found in the media supernatants of the co-cultures. Conclusions The co-culture system described here is suitable for testing neuronal differentiation capability of numerous types of stem cells. Especially in the case of human cells, it may be of clinical relevance for future cell-based therapeutic applications.
Collapse
Affiliation(s)
- Anna E. Petschnik
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | - Benjamin Fell
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | - Stephan Tiede
- Department of Dermatology, Allergology and Venerology, University of Lübeck, Lübeck, Germany
| | | | - Ralph Pries
- ENT Department, University of Lübeck, Lübeck, Germany
| | - Charli Kruse
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | - Sandra Danner
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
- * E-mail:
| |
Collapse
|
26
|
Alexanian AR, Fehlings MG, Zhang Z, Maiman DJ. Transplanted neurally modified bone marrow-derived mesenchymal stem cells promote tissue protection and locomotor recovery in spinal cord injured rats. Neurorehabil Neural Repair 2011; 25:873-80. [PMID: 21844281 DOI: 10.1177/1545968311416823] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Stem cell-based therapy for repair and replacement of lost neural cells is a promising treatment for central nervous system (CNS) diseases. Bone marrow (BM)-derived mesenchymal stem cells (MSCs) can differentiate into neural phenotypes and be isolated and expanded for autotransplantation with no risk of rejection. OBJECTIVE The authors examined whether transplanted neurally induced human MSCs (NI hMSCs), developed by a new procedure, can survive, differentiate, and promote tissue protection and functional recovery in injured spinal cord (ISC) rats. METHODS Neural induction was achieved by exposing cells simultaneously to inhibitors of DNA methylation, histone deacetylation, and pharmacological agents that increased cAMP levels. Three groups of adult female Sprague-Dawley rats were injected immediately rostral and caudal to the midline lesion with phosphate-buffered saline, MSCs, or NI hMSCs, 1 week after a spinal cord impact injury at T-8. Functional outcome was measured using the Basso Beattie Bresnahan (BBB) locomotor rating scale and thermal sensitivity test on a weekly basis up to 12 weeks postinjury. Graft integration and anatomy of spinal cord was assessed by stereological, histochemical, and immunohistochemical techniques. RESULTS The transplanted NI hMSCs survived, differentiated, and significantly improved locomotor recovery of ISC rats. Transplantation also reduced the volume of lesion cavity and white matter loss. CONCLUSION This method of hMSC modification may provide an alternative source of autologous adult stem cells for CNS repair.
Collapse
|