1
|
Gamit N, Patil M, Soumya BS, Dharmarajan A, Warrier S. Development of In Vitro Parkinson's Disease Model Mediated by MPP+ and α-Synuclein Using Wharton's Jelly Mesenchymal Stem Cells. CNS Neurosci Ther 2025; 31:e70299. [PMID: 40260646 PMCID: PMC12012574 DOI: 10.1111/cns.70299] [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: 09/28/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 04/23/2025] Open
Abstract
MAIN PROBLEM The mechanism behind Parkinson's disease (PD) is still unclear, and a cure to stop its progression is yet to be found. This is mainly due to the lack of effective human PD models. To address this, we generated an in vitro PD model using Wharton's jelly-derived mesenchymal stem cells (WJMSCs). METHODS WJMSCs were isolated from the umbilical cord using an enzymatic method. MSCs were characterized by RT-PCR, immunofluorescence, and trilineage differentiation. MSCs were differentiated into dopaminergic neuron-like cells (DAN) and further degenerated by treating them with either MPP+ iodide or the A53T mutated α-synuclein variant. Gene expression analysis by qRT-PCR and protein analysis by immunofluorescence, flow cytometry, and ELISA were performed. Assays to measure LDH, ROS, NO, GSH, and mitochondrial membrane potential were also performed after degeneration. RESULTS WJMSCs were positive for MSC markers and were able to differentiate into adipocytes, chondrocytes, and osteocytes. DAN obtained after the differentiation of WJMSCs for 48 h expressed neuronal markers such as synapsin 1, neuropilin, neurofilament, and MAPT along with dopaminergic markers such as Nurr1, DAT, TH, DDC, and KCNJ6 and were functionally active. Upon degeneration of DAN by MPP+ or A53T, elevated levels of SNCA and downregulation of TH, Nurr1, DAT, and KCNJ6 were observed. Furthermore, increased expression of α-SYN was detected at the protein level as well. Finally, reduction in mitochondrial membrane potential and GSH levels along with an increase in intracellular ROS, nitrite production, and LDH levels confirmed that the in vitro PD-like model exhibited the molecular characteristics of PD. CONCLUSION This model is rapid, cost-efficient, and effective for understanding the molecular mechanisms of the disease and can also be used for screening of emerging therapeutics for PD.
Collapse
Affiliation(s)
- Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal Academy of Higher Education (MAHE)BangaloreIndia
| | - Manasi Patil
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal Academy of Higher Education (MAHE)BangaloreIndia
| | - B. S. Soumya
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal Academy of Higher Education (MAHE)BangaloreIndia
| | - Arun Dharmarajan
- School of Human SciencesThe University of Western AustraliaNedlandsWestern AustraliaAustralia
- Curtin Medical SchoolCurtin UniversityPerthWestern AustraliaAustralia
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal Academy of Higher Education (MAHE)BangaloreIndia
- Department of Biotechnology, Faculty of Biomedical Sciences and TechnologySri Ramachandra Institute of Higher Education and ResearchChennaiIndia
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Mesenchymal Stromal Cells Preconditioning: A New Strategy to Improve Neuroprotective Properties. Int J Mol Sci 2022; 23:ijms23042088. [PMID: 35216215 PMCID: PMC8878691 DOI: 10.3390/ijms23042088] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Neurological diseases represent one of the main causes of disability in human life. Consequently, investigating new strategies capable of improving the quality of life in neurological patients is necessary. For decades, researchers have been working to improve the efficacy and safety of mesenchymal stromal cells (MSCs) therapy based on MSCs’ regenerative and immunomodulatory properties and multilinear differentiation potential. Therefore, strategies such as MSCs preconditioning are useful to improve their application to restore damaged neuronal circuits following neurological insults. This review is focused on preconditioning MSCs therapy as a potential application to major neurological diseases. The aim of our work is to summarize both the in vitro and in vivo studies that demonstrate the efficacy of MSC preconditioning on neuronal regeneration and cell survival as a possible application to neurological damage.
Collapse
|
5
|
Rosich K, Hanna BF, Ibrahim RK, Hellenbrand DJ, Hanna A. The Effects of Glial Cell Line-Derived Neurotrophic Factor after Spinal Cord Injury. J Neurotrauma 2017; 34:3311-3325. [DOI: 10.1089/neu.2017.5175] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Konstantin Rosich
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| | - Bishoy F. Hanna
- Department of Neurological Surgery, Ross University School of Medicine, Dominica, West Indies
| | - Rami K. Ibrahim
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| | - Daniel J. Hellenbrand
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin
| | - Amgad Hanna
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
6
|
Zheng Y, Huang C, Liu F, Lin H, Yang X, Zhang Z. Comparison of the neuronal differentiation abilities of bone marrow‑derived and adipose tissue‑derived mesenchymal stem cells. Mol Med Rep 2017; 16:3877-3886. [PMID: 28731172 PMCID: PMC5646965 DOI: 10.3892/mmr.2017.7069] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/17/2017] [Indexed: 11/22/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) and adipose tissue-derived mesenchymal stem cells (ADSCs) are able to differentiate into neuron-like cells when exposed to small molecule compounds, however the specific differences in their neuronal differentiation abilities remain to be fully elucidated. The present study aimed to compare the neuronal differentiation abilities of BMSCs and ADSCs. BMSCs and ADSCs from the same Sprague Dawley rats were isolated and cultured for use. The proliferation capacity was revealed using a cell counting method. Following BMSCs and ADSCs induction by four types of small-molecular compounds, the expression of various neuronal markers and the secretion of several neurotrophic factors were detected by immunofluorescence, western blotting, reverse transcription-quantitative polymerase chain reaction and ELISA. It was demonstrated that the ADSCs exhibited an increased proliferation capacity compared with BMSCs, according to cumulative population doubling analyses. Following a 7-day neuronal induction period, BMSCs and ADSCs exhibited a neuron-like morphology, and were termed neuronal induced (NI)-BMSCs and NI-ADSCs. They expressed neuronal markers including β-tubulin III, microtubule associated protein 2 and choline acetyltransferase. The number of NI-BMSCs that positively expressed the neuronal markers was significantly decreased compared with NI-ADSCs, and the expression and secretion of the neurotrophic factors nerve growth factor and 3′-nucleotidase in NI-BMSCs were additionally decreased compared with NI-ADSCs. The findings of the present study indicated that the neuronal differentiation abilities and neurotrophic factor secretion abilities of ADSCs were increased compared with BMSCs. ADSCs may therefore act as efficient candidates in cell transplantation therapy for diseases and injuries of the nervous system.
Collapse
Affiliation(s)
- Yani Zheng
- Department of Anatomy, Institute of Biomedical Engineering, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Chao Huang
- Department of Anatomy, Institute of Biomedical Engineering, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Fang Liu
- Department of Anatomy, Institute of Biomedical Engineering, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Haiyan Lin
- Department of Anatomy, Institute of Biomedical Engineering, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Xiangqun Yang
- Department of Anatomy, Institute of Biomedical Engineering, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Zhiying Zhang
- Department of Anatomy, Institute of Biomedical Engineering, The Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
7
|
Zhao C, Li H, Zhao XJ, Liu ZX, Zhou P, Liu Y, Feng MJ. Heat shock protein 60 affects behavioral improvement in a rat model of Parkinson’s disease grafted with human umbilical cord mesenchymal stem cell-derived dopaminergic-like neurons. Neurochem Res 2016; 41:1238-49. [DOI: 10.1007/s11064-015-1816-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 11/19/2015] [Accepted: 12/23/2015] [Indexed: 01/01/2023]
|
8
|
Nakano R, Edamura K, Nakayama T, Narita T, Okabayashi K, Sugiya H. Fibroblast Growth Factor Receptor-2 Contributes to the Basic Fibroblast Growth Factor-Induced Neuronal Differentiation in Canine Bone Marrow Stromal Cells via Phosphoinositide 3-Kinase/Akt Signaling Pathway. PLoS One 2015; 10:e0141581. [PMID: 26523832 PMCID: PMC4629880 DOI: 10.1371/journal.pone.0141581] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/09/2015] [Indexed: 11/26/2022] Open
Abstract
Bone marrow stromal cells (BMSCs) are considered as candidates for regenerative therapy and a useful model for studying neuronal differentiation. The role of basic fibroblast growth factor (bFGF) in neuronal differentiation has been previously studied; however, the signaling pathway involved in this process remains poorly understood. In this study, we investigated the signaling pathway in the bFGF-induced neuronal differentiation of canine BMSCs. bFGF induced the mRNA expression of the neuron marker, microtubule associated protein-2 (MAP2) and the neuron-like morphological change in canine BMSCs. In the presence of inhibitors of fibroblast growth factor receptors (FGFR), phosphatidylinositol 3-kinase (PI3K) and Akt, i.e., SU5402, LY294002, and MK2206, respectively, bFGF failed to induce the MAP2 mRNA expression and the neuron-like morphological change. bFGF induced Akt phosphorylation, but it was attenuated by the FGFR inhibitor SU5402 and the PI3K inhibitor LY294002. In canine BMSCs, expression of FGFR-1 and FGFR-2 was confirmed, but only FGFR-2 activation was detected by cross-linking and immunoprecipitation analysis. Small interfering RNA-mediated knockdown of FGFR-2 in canine BMSCs resulted in the attenuation of bFGF-induced Akt phosphorylation. These results suggest that the FGFR-2/PI3K/Akt signaling pathway is involved in the bFGF-induced neuronal differentiation of canine BMSCs.
Collapse
Affiliation(s)
- Rei Nakano
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Kazuya Edamura
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Tomohiro Nakayama
- Laboratory of Veterinary Radiology, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Takanori Narita
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Ken Okabayashi
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
| | - Hiroshi Sugiya
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Kameino, Fujisawa, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
9
|
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
|
10
|
Liu X, Huang Q, Li F, Li CY. Enhancing the efficiency of direct reprogramming of human primary fibroblasts into dopaminergic neuron-like cells through p53 suppression. SCIENCE CHINA-LIFE SCIENCES 2014; 57:867-75. [PMID: 25129808 DOI: 10.1007/s11427-014-4730-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 06/28/2014] [Indexed: 12/23/2022]
Abstract
Dopaminergic (DA) neuron-like cells obtained through direct reprogramming of primary human fibroblasts offer exciting opportunities for treatment of Parkinson's disease. A significant obstacle is the low efficiency of conversion during the reprogramming process. Here, we demonstrate that the suppression of p53 significantly enhances the efficiency of transcription factor-mediated conversion of human fibroblasts into functional dopaminergic neurons. In particular, blocking p53 activity using a dominant-negative p53 (p53-DN) in IMR90 cells increases the conversion efficiency by 5-20 fold. The induced DA neuron-like cells exhibit dopamine neuron-specific gene expression, significant dopamine uptake and production capacities, and enables symptomatic relief in a rat Parkinson's disease model. Taken together, our findings suggest that p53 is a critical barrier in direct reprogramming of fibroblast into dopaminergic neurons.
Collapse
Affiliation(s)
- XinJian Liu
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | | | | | | |
Collapse
|
11
|
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
|
12
|
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
|