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
|
Wei C, Yu P, Cheng L. Hematopoietic Reprogramming Entangles with Hematopoiesis. Trends Cell Biol 2020; 30:752-763. [PMID: 32861580 DOI: 10.1016/j.tcb.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023]
Abstract
Hematopoiesis generally refers to hematopoietic development in fetuses and adults, as well as to hematopoietic stem cell differentiation into progeny lineages. The multiple processes that generate diverse hematopoietic cells have been considered to be unidirectional. However, many reports have recently demonstrated that these processes are not only reversible but also interconvertible via cell reprogramming. The cell reprogramming that occurs in hematopoietic cells is termed hematopoietic reprogramming. We focus on both autogenous and artificial hematopoietic reprogramming under physiological and pathological conditions that is mainly directed by the actions of transcription factors (TFs), chemical compounds, or extracellular cytokines. A comprehensive understanding of hematopoietic reprogramming will help us not only to generate desirable cells for cell therapy but also to further analyze normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Chuijin Wei
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Pei Yu
- Department of Orthopaedics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
3
|
Ozkul Y, Galderisi U. The Impact of Epigenetics on Mesenchymal Stem Cell Biology. J Cell Physiol 2016; 231:2393-2401. [PMID: 26960183 DOI: 10.1002/jcp.25371] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/07/2016] [Indexed: 02/06/2023]
Abstract
Changes in epigenetic marks are known to be important regulatory factors in stem cell fate determination and differentiation. In the past years, the investigation of the epigenetic regulation of stem cell biology has largely focused on embryonic stem cells (ESCs). Contrarily, less is known about the epigenetic control of gene expression during differentiation of adult stem cells (AdSCs). Among AdSCs, mesenchymal stem cells (MSCs) are the most investigated stem cell population because of their enormous potential for therapeutic applications in regenerative medicine and tissue engineering. In this review, we analyze the main studies addressing the epigenetic changes in MSC landscape during in vitro cultivation and replicative senescence, as well as follow osteocyte, chondrocyte, and adipocyte differentiation. In these studies, histone acetylation, DNA methylation, and miRNA expression are among the most investigated phenomena. We describe also epigenetic changes that are associated with in vitro MSC trans-differentiation. Although at the at initial stage, the epigenetics of MSCs promise to have profound implications for stem cell basic and applied research. J. Cell. Physiol. 231: 2393-2401, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yusuf Ozkul
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | - Umberto Galderisi
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| |
Collapse
|
4
|
Valproic acid enhances neuronal differentiation of sympathoadrenal progenitor cells. Mol Psychiatry 2015; 20:941-50. [PMID: 25707399 DOI: 10.1038/mp.2015.3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 11/28/2014] [Accepted: 12/19/2014] [Indexed: 02/08/2023]
Abstract
The antiepileptic drug valproic acid (VPA) has been shown to influence the neural differentiation and neurite outgrowth of neural stem cells. Sympathoadrenal progenitor cells share properties with neural stem cells and are considered a potential cell source in the treatment of neurodegenerative diseases. The present study therefore aims at modulating the neural differentiation potential of these cells by treatment with the histone deacetylase inhibitor VPA. We studied the epigenetic effects of VPA in two culture conditions: suspension conditions aimed to expand adrenomedullary sympathoadrenal progenitors within free-floating chromospheres and adherent cell cultures optimized to derive neurons. Treatment of chromospheres with VPA may launch neuronal differentiation mechanisms and improve their neurogenic potential upon transplantation. However, also transplantation of differentiated functional neurons could be beneficial. Treating chromospheres for 7 days with clinically relevant concentrations of VPA (2 mm) revealed a decrease of neural progenitor markers Nestin, Notch2 and Sox10. Furthermore, VPA initiated catecholaminergic neuronal differentiation indicated by upregulation of the neuronal marker β-III-tubulin, the dopaminergic transcription factor Pitx3 and the catecholaminergic enzymes TH and GTPCH. In adherent neural differentiation conditions, VPA treatment improved the differentiation of sympathoadrenal progenitor cells into catecholaminergic neurons with significantly elevated levels of nor- and epinephrine. In conclusion, similar to neural stem cells, VPA launches differentiation mechanisms in sympathoadrenal progenitor cells that result in increased generation of functional neurons. Thus, data from this study will be relevant to the potential use of chromaffin progenitors in transplantation therapies of neurodegenerative diseases.
Collapse
|
5
|
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
|
6
|
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
|
7
|
Mukhopadhyay A. Perspective on liver regeneration by bone marrow-derived stem cells-a scientific realization or a paradox. Cytotherapy 2013; 15:881-92. [PMID: 23623692 DOI: 10.1016/j.jcyt.2013.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/27/2012] [Accepted: 02/09/2013] [Indexed: 12/11/2022]
Abstract
Bone marrow (BM)-derived stem cells are reported to have cellular plasticity, which provoked many investigators to use of these cells in the regeneration of nonhematopoietic tissues. However, adult stem cell plasticity contradicts our classic understanding on progressive restriction of the developmental potential of a cell type. Many alternate mechanisms have been proposed to explain this phenomenon; the working hypotheses for elucidating the cellular plasticity of BM-derived stem cells are on the basis of direct differentiation and/or fusion between donor and recipient cells. This review dissects the different outcomes of the investigations on liver regeneration, which were performed with the use of BM-derived stem cells in experimental animals, and reveals some critical factors to explain cellular plasticity. It has been hypothesized that the competent BM-derived stem/progenitor cells, under the influence of liver-regenerating cues, can directly differentiate into hepatic cells. This differentiation takes place as a result of genetic reprogramming, which may be possible in the chemically induced acute liver injury model or at the stage of fetal liver development. Cellular plasticity emerges as an important phenomenon in cell-based therapies for the treatment of many liver diseases in which tissue regeneration is necessary.
Collapse
Affiliation(s)
- Asok Mukhopadhyay
- Stem Cell Biology & Center for Molecular Medicine, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India.
| |
Collapse
|
8
|
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
|
9
|
Transdifferentiation: a cell and molecular reprogramming process. Cell Tissue Res 2012; 348:379-96. [PMID: 22526624 DOI: 10.1007/s00441-012-1403-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 03/01/2012] [Indexed: 12/13/2022]
Abstract
Evidence has emerged recently indicating that differentiation is not entirely a one-way process, and that it is possible to convert one cell type to another, both in vitro and in vivo. This phenomenon is called transdifferentiation, and is generally defined as the stable switch of one cell type to another. Transdifferentiation plays critical roles during development and in regeneration pathways in nature. Although this phenomenon occurs rarely in nature, recent studies have been focused on transdifferentiation and the reprogramming ability of cells to produce specific cells with new phenotypes for use in cell therapy and regenerative medicine. Thus, understanding the principles and the mechanism of this process is important for producing desired cell types. Here some well-documented examples of transdifferentiation, and their significance in development and regeneration are reviewed. In addition, transdifferentiation pathways are considered and their potential molecular mechanisms, especially the role of master switch genes, are considered. Finally, the significance of transdifferentiation in regenerative medicine is discussed.
Collapse
|
10
|
Expression of α2, α5 and α6 subunits of integrin in de-differentiated NIH3T3 cells by cell-free extract of embryonic stem cells. Mol Biol Rep 2012; 39:7339-46. [DOI: 10.1007/s11033-012-1565-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 01/25/2012] [Indexed: 12/22/2022]
|
11
|
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
|
12
|
Obier N, Uhlemann CF, Müller AM. Inhibition of histone deacetylases by Trichostatin A leads to a HoxB4-independent increase of hematopoietic progenitor/stem cell frequencies as a result of selective survival. Cytotherapy 2011; 12:899-908. [PMID: 20210674 DOI: 10.3109/14653240903580254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND DNA and chromatin modifications are critical mediators in the establishment and maintenance of cell type-specific gene expression patterns that constitute cellular identities. One type of modification, the acetylation and deacetylation of histones, occurs reversibly on lysine ε-NH₃(+) groups of core histones via histone acetyl transferases (HAT) and histone deacetylases (HDAC). Hyperacetylated histones are associated with active chromatin domains, whereas hypoacetylated histones are enriched in non-transcribed loci. METHODS We analyzed global histone H4 acetylation and HDAC activity levels in mature lineage marker-positive (Lin(+)) and progenitor lineage marker-negative (Lin⁻) hematopoietic cells from murine bone marrow (BM). In addition, we studied the effects of HDAC inhibition on hematopoietic progenitor/stem cell (HPSC) frequencies, cell survival, differentiation and HoxB4 dependence. RESULTS We observed that Lin⁻ and Lin(+) cells do not differ in global histone H4 acetylation but in HDAC activity levels. Further, we saw that augmented histone acetylation achieved by transient Trichostatin A (TSA) treatment increased the frequency of cells with HPSC immunophenotype and function in the heterogeneous pool of BM cells. Induction of histone hyperacetylation in differentiated BM cells was detrimental, as evidenced by preferential death of mature BM cells upon HDAC inhibition. Finally, TSA treatment of BM cells from HoxB4(-/-) mice revealed that the HDAC inhibitor-mediated increase in HPSC frequencies was independent of HoxB4. CONCLUSIONS Overall, these data indicate the potential of chromatin modifications for the regulation of HPSC. Chromatin-modifying agents may provide potential strategies for ex vivo expansion of HPSC.
Collapse
Affiliation(s)
- Nadine Obier
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), Universität Würzburg, Würzburg, Germany
| | | | | |
Collapse
|
13
|
Alexanian AR. An efficient method for generation of neural-like cells from adult human bone marrow-derived mesenchymal stem cells. Regen Med 2011; 5:891-900. [PMID: 21082889 DOI: 10.2217/rme.10.67] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Stem cell-based therapies to repair and replace lost neural cells are a highly promising treatment for CNS diseases. Bone marrow (BM)-derived mesenchymal stem cells (MSCs) have great potential as therapeutic agents against neurological maladies, since they have the ability to differentiate into neural phenotypes and can be readily isolated and expanded for autotransplantation with no risk of rejection. In our previous studies, we demonstrated that neural cells could be efficiently generated from mouse BM-derived MSCs by exposing cells to epigenetic modifiers and a neural environment. The main idea of this approach was the reactivation of pluripotency-associated genes in MSCs before exposing them to neural-inducing factors. AIM In this study, we used a similar approach to efficiently generate neural cells from human BM-derived MSCs. METHOD Neural induction was achieved by exposing cells simultaneously to inhibitors of DNA methylation and histone deacetylation, and pharmacological agents that increase cAMP levels. RESULTS The expression of pluripotency and neural markers was confirmed with immunocytochemistry, western blot and real-time PCR. ELISA studies showed that these neurally induced-human MSCs cells released the neurotrophic factors glial cell-derived neurotrophic factor and brain-derived neurotrophic factor. CONCLUSION Human MSCs that are neurally modified with this methodology could be a useful source of cells for CNS repair and regeneration.
Collapse
Affiliation(s)
- Arshak R Alexanian
- Neuroscience Research Laboratories, Department of Neurosurgery, VA Medical Center - Research 151, 5000 West National Avenue, Milwaukee, Wisconsin, WI 53295, USA.
| |
Collapse
|
14
|
Lee SH, Appleby V, Jeyapalan JN, Palmer RD, Nicholson JC, Sottile V, Gao E, Coleman N, Scotting PJ. Variable methylation of the imprinted gene, SNRPN, supports a relationship between intracranial germ cell tumours and neural stem cells. J Neurooncol 2011; 101:419-28. [PMID: 20582452 DOI: 10.1007/s11060-010-0275-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 06/16/2010] [Indexed: 12/14/2022]
Abstract
Germ cell tumours (GCTs) are a diverse group of neoplasms all of which are generally believed to arise from germ cell progenitors (PGCs). Even those that form in the nervous system are likewise believed to be PGC-derived, despite being found a great distance from the normal location of germ cells. The primary evidence in favour of this model for the origins of intracranial GCTs is that they share molecular features with other GCTs. Those features include shared gene expression and a lack of methylation of imprinted genes, including SNRPN. Contrary to this model, we have proposed that endogenous neural stem cells of the brain are a more likely origin for these tumours. We show here that the lack of methylation of SNRPN that has previously been taken to indicate an origin for GCTs from PGCs is also seen in neural stem cells of mice and humans. We believe that, in the light of these and other recent observations, endogenous neural precursors of the brain are a more plausible origin for intracranial GCTs than are misplaced PGCs.
Collapse
Affiliation(s)
- Shih-Han Lee
- Children's Brain Tumour Research Centre, Institute of Genetics, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Godmann M, May E, Kimmins S. Epigenetic mechanisms regulate stem cell expressed genes Pou5f1 and Gfra1 in a male germ cell line. PLoS One 2010; 5:e12727. [PMID: 20856864 PMCID: PMC2939054 DOI: 10.1371/journal.pone.0012727] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 08/19/2010] [Indexed: 01/15/2023] Open
Abstract
Male fertility is declining and an underlying cause may be due to environment-epigenetic interactions in developing sperm, yet nothing is known of how the epigenome controls gene expression in sperm development. Histone methylation and acetylation are dynamically regulated in spermatogenesis and are sensitive to the environment. Our objectives were to determine how histone H3 methylation and acetylation contribute to the regulation of key genes in spermatogenesis. A germ cell line, GC-1, was exposed to either the control, or the chromatin modifying drugs tranylcypromine (T), an inhibitor of the histone H3 demethylase KDM1 (lysine specific demethylase 1), or trichostatin (TSA), an inhibitor of histone deacetylases, (HDAC). Quantitative PCR (qPCR) was used to identify genes that were sensitive to treatment. As a control for specificity the Myod1 (myogenic differentiation 1) gene was analyzed. Chromatin immunoprecipitation (ChIP) followed by qPCR was used to measure histone H3 methylation and acetylation at the promoters of target genes and the control, Myod1. Remarkably, the chromatin modifying treatment specifically induced the expression of spermatogonia expressed genes Pou5f1 and Gfra1. ChIP-qPCR revealed that induction of gene expression was associated with a gain in gene activating histone H3 methylation and acetylation in Pou5f1 and Gfra1 promoters, whereas CpG DNA methylation was not affected. Our data implicate a critical role for histone H3 methylation and acetylation in the regulation of genes expressed by spermatogonia – here, predominantly mediated by HDAC-containing protein complexes.
Collapse
Affiliation(s)
- Maren Godmann
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Erin May
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
| | - Sarah Kimmins
- Department of Animal Science, McGill University, Ste-Anne-de-Bellevue, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
16
|
Zhuang J, Ye Y, Liu X, Li F, Pan X, Chen Z, Luo H, Ge Y, Ge J, Kaminski J, Yu K. DNA demethylation in retinal neurocytes contributes to the upregulation of DNA repair protein, Ku80. Neuroreport 2010; 21:282-6. [DOI: 10.1097/wnr.0b013e328336ee7e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
17
|
Artaza JN, Sirad F, Ferrini MG, Norris KC. 1,25(OH)2vitamin D3 inhibits cell proliferation by promoting cell cycle arrest without inducing apoptosis and modifies cell morphology of mesenchymal multipotent cells. J Steroid Biochem Mol Biol 2010; 119:73-83. [PMID: 20064609 PMCID: PMC2828517 DOI: 10.1016/j.jsbmb.2010.01.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2009] [Revised: 01/02/2010] [Accepted: 01/04/2010] [Indexed: 01/09/2023]
Abstract
The vitamin D receptor (VDR) and its ligand 1,25D play an important role in regulating cell growth and cell fate. We examined the effect of 1,25D on cell morphology, cell proliferation, cell cycle progression and apoptosis on mesenchymal multipotent cells. Multipotent cells were treated with and without 1,25D in a time- and dose-dependent manner. Changes in cell morphology were evaluated by a green fluorescence fluorocrome. Cell proliferation was determined by the Formazan assay and PCNA antigen expression. The expression of genes related to the cell cycle was analyzed by DNA microarrays, RT(2)PCR arrays and western blots. Apoptosis was evaluated by TUNEL assay, and the expression of pro- and anti-apoptotic related genes by RT(2)PCR arrays and western blots. 1,25D inhibited cell proliferation, induced cell cycle arrest, and promoted accumulation of cells in G0/G1 phase without inducing apoptosis. An increase in cell size was associated with a decrease in the GTPase Rho and the atypical Rho family GTPase Rhou/Wrch-1 expression without inducing Wnt-1 expression. Survivin expression was also increased and may represent a novel 1,25D-mediated pathway regulating tissue injury and fibrosis. The data provide a mechanistic explanation for the anti-proliferative and anti-apoptotic properties of 1,25D in mesenchymal multipotent cells.
Collapse
Affiliation(s)
- Jorge N Artaza
- Department of Internal Medicine, Charles Drew University of Medicine & Science, Los Angeles, CA 90059, USA.
| | | | | | | |
Collapse
|
18
|
Vukicevic V, Jauch A, Dinger TC, Gebauer L, Hornich V, Bornstein SR, Ehrhart-Bornstein M, Müller AM. Genetic instability and diminished differentiation capacity in long-term cultured mouse neurosphere cells. Mech Ageing Dev 2010; 131:124-32. [DOI: 10.1016/j.mad.2010.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 12/20/2009] [Accepted: 01/06/2010] [Indexed: 12/25/2022]
|
19
|
Takehara T, Teramura T, Onodera Y, Kishigami S, Matsumoto K, Saeki K, Fukuda K, Hosoi Y. Potential Existence of Stem Cells With Multiple Differentiation Abilities to Three Different Germ Lineages in Mouse Neurospheres. Stem Cells Dev 2009; 18:1433-40. [DOI: 10.1089/scd.2008.0239] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Toshiyuki Takehara
- Graduate School of Biology-Oriented Science and Technology, Kinki University, Nishimitani, Kinokawa, Wakayama, Japan
| | - Takeshi Teramura
- Institute of Advanced Clinical Medicine, Kinki University School of Medicine, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Yuta Onodera
- Institute of Advanced Clinical Medicine, Kinki University School of Medicine, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Satoshi Kishigami
- Graduate School of Biology-Oriented Science and Technology, Kinki University, Nishimitani, Kinokawa, Wakayama, Japan
| | - Kazuya Matsumoto
- Graduate School of Biology-Oriented Science and Technology, Kinki University, Nishimitani, Kinokawa, Wakayama, Japan
| | - Kazuhiro Saeki
- Graduate School of Biology-Oriented Science and Technology, Kinki University, Nishimitani, Kinokawa, Wakayama, Japan
| | - Kanji Fukuda
- Institute of Advanced Clinical Medicine, Kinki University School of Medicine, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
- Department of Rehabilitation, Kinki University School of Medicine, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Yoshihiko Hosoi
- Graduate School of Biology-Oriented Science and Technology, Kinki University, Nishimitani, Kinokawa, Wakayama, Japan
| |
Collapse
|
20
|
Snykers S, De Kock J, Rogiers V, Vanhaecke T. In vitro differentiation of embryonic and adult stem cells into hepatocytes: state of the art. Stem Cells 2009; 27:577-605. [PMID: 19056906 PMCID: PMC2729674 DOI: 10.1634/stemcells.2008-0963] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stem cells are a unique source of self-renewing cells within the human body. Before the end of the last millennium, adult stem cells, in contrast to their embryonic counterparts, were considered to be lineage-restricted cells or incapable of crossing lineage boundaries. However, the unique breakthrough of muscle and liver regeneration by adult bone marrow stem cells at the end of the 1990s ended this long-standing paradigm. Since then, the number of articles reporting the existence of multipotent stem cells in skin, neuronal tissue, adipose tissue, and bone marrow has escalated, giving rise, both in vivo and in vitro, to cell types other than their tissue of origin. The phenomenon of fate reprogrammation and phenotypic diversification remains, though, an enigmatic and rare process. Understanding how to control both proliferation and differentiation of stem cells and their progeny is a challenge in many fields, going from preclinical drug discovery and development to clinical therapy. In this review, we focus on current strategies to differentiate embryonic, mesenchymal(-like), and liver stem/progenitor cells into hepatocytes in vitro. Special attention is paid to intracellular and extracellular signaling, genetic modification, and cell-cell and cell-matrix interactions. In addition, some recommendations are proposed to standardize, optimize, and enrich the in vitro production of hepatocyte-like cells out of stem/progenitor cells.
Collapse
Affiliation(s)
- Sarah Snykers
- Department of Toxicology, Vrije Universiteit Brussel, Belgium.
| | | | | | | |
Collapse
|
21
|
Snykers S, Henkens T, De Rop E, Vinken M, Fraczek J, De Kock J, De Prins E, Geerts A, Rogiers V, Vanhaecke T. Role of epigenetics in liver-specific gene transcription, hepatocyte differentiation and stem cell reprogrammation. J Hepatol 2009; 51:187-211. [PMID: 19457566 DOI: 10.1016/j.jhep.2009.03.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Controlling both growth and differentiation of stem cells and their differentiated somatic progeny is a challenge in numerous fields, from preclinical drug development to clinical therapy. Recently, new insights into the underlying molecular mechanisms have unveiled key regulatory roles of epigenetic marks driving cellular pluripotency, differentiation and self-renewal/proliferation. Indeed, the transcription of genes, governing cell-fate decisions during development and maintenance of a cell's differentiated status in adult life, critically depends on the chromatin accessibility of transcription factors to genomic regulatory and coding regions. In this review, we discuss the epigenetic control of (liver-specific) gene-transcription and the intricate interplay between chromatin modulation, including histone (de)acetylation and DNA (de)methylation, and liver-enriched transcription factors. Special attention is paid to their role in directing hepatic differentiation of primary hepatocytes and stem cells in vitro.
Collapse
Affiliation(s)
- Sarah Snykers
- Department of Toxicology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Chen M, Takano-Maruyama M, Pereira-Smith OM, Gaufo GO, Tominaga K. MRG15, a component of HAT and HDAC complexes, is essential for proliferation and differentiation of neural precursor cells. J Neurosci Res 2009; 87:1522-31. [PMID: 19115414 PMCID: PMC2913448 DOI: 10.1002/jnr.21976] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Neurogenesis during development depends on the coordinated regulation of self-renewal and differentiation of neural precursor cells (NPCs). Chromatin regulation is a key step in self-renewal activity and fate decision of NPCs. However, the molecular mechanism or mechanisms of this regulation is not fully understood. Here, we demonstrate for the first time that MRG15, a chromatin regulator, is important for proliferation and neural fate decision of NPCs. Neuroepithelia from Mrg15-deficient embryonic brain are much thinner than those from control, and apoptotic cells increase in this region. We isolated NPCs from Mrg15-deficient and wild-type embryonic whole brains and produced neurospheres to measure the self-renewal and differentiation abilities of these cells in vitro. Neurospheres culture from Mrg15-deficient embryo grew less efficiently than those from wild type. Measurement of proliferation by means of BrdU (bromodeoxyuridine) incorporation revealed that Mrg15-deficient NPCs have reduced proliferation ability and apoptotic cells do not increase during in vitro culture. The reduced proliferation of Mrg15-deficient NPCs most likely accounts for the thinner neuroepithelia in Mrg15-deficient embryonic brain. Moreover, we also demonstrate Mrg15-deficient NPCs are defective in differentiation into neurons in vitro. Our results demonstrate that MRG15 has more than one function in neurogenesis and defines a novel role for this chromatin regulator that integrates proliferation and cell-fate determination in neurogenesis during development.
Collapse
Affiliation(s)
- Meizhen Chen
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, 15535 Lambda Drive, STCBM #3.100, San Antonio, TX 78245, USA
| | - Masumi Takano-Maruyama
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, BSB 2.03.42, San Antonio, TX 78249, USA
| | - Olivia M. Pereira-Smith
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, 15535 Lambda Drive, STCBM #3.100, San Antonio, TX 78245, USA
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, 15535 Lambda Drive, STCBM #3.100, San Antonio, TX 78245, USA
| | - Gary O. Gaufo
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, BSB 2.03.42, San Antonio, TX 78249, USA
| | - Kaoru Tominaga
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, 15535 Lambda Drive, STCBM #3.100, San Antonio, TX 78245, USA
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, 15535 Lambda Drive, STCBM #3.100, San Antonio, TX 78245, USA
| |
Collapse
|
23
|
Lederer CW, Santama N. Neural stem cells: mechanisms of fate specification and nuclear reprogramming in regenerative medicine. Biotechnol J 2009; 3:1521-38. [PMID: 19072908 DOI: 10.1002/biot.200800193] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently, intense interest in the potential use of neural stem cells (NSC) in the clinical therapy of brain disease and injury has resulted in rapid progress in research on the properties of NSC, their innate and directed differentiation potential and the induced reprogramming of differentiated somatic cells to revert to a pluripotent NSC-like state. The aim of this review is to give an overview of our current operational definitions of the NSC lineage, the growing understanding of extrinsic and intrinsic mechanisms, including heritable but reversible epigenetic chromatin modifications that regulate the maintenance and differentiation of NSC in vivo, and to emphasize ground-breaking efforts of cellular reprogramming with the view to generating patient-specific stem cells for cell replacement therapy. This is set against a summary of current practical procedures for the isolation, research and application of NSC, and of the state of the art in NSC-based regenerative medicine of the nervous system. Both provide the backdrop for the translation of recent findings into innovative clinical applications, with the hope of increasing the safety, efficiency and ethical acceptability of NSC-based therapies in the near future.
Collapse
|
24
|
Artaza JN, Norris KC. Vitamin D reduces the expression of collagen and key profibrotic factors by inducing an antifibrotic phenotype in mesenchymal multipotent cells. J Endocrinol 2009; 200:207-21. [PMID: 19036760 PMCID: PMC3787314 DOI: 10.1677/joe-08-0241] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypovitaminosis D is an important public health problem. Serum 25-hydroxyvitamin D (25-OHD) is now recognized as an independent predictor for cardiovascular and related diseases (CVD) as well as other chronic medical conditions. However, the biologic pathways through which these effects are mediated remain poorly understood. We hypothesized that exposing mesenchymal multipotent cells (MMCs) to the active form of vitamin D would increase the expression of selected antifibrotic factors that in turn would ameliorate the progression of chronic diseases. MMCs were primed with 5'-azacytidine to induce a fibrotic phenotype and then treated with active vitamin D (1,25D) or ethanol <0.1% as vehicle in a time course manner (30 min, 1, 5, and 24 h, and for 4 and 7 days). The addition of 1,25D to MMCs promotes: a) increased expression and nuclear translocation of the vitamin D receptor; b) decreased expression of TGFB1 and plasminogen activator inhibitor (SERPINE1), two well-known profibrotic factors; c) decreased expression of collagen I, III and other collagens isoforms; and d) increased expression of several antifibrotic factors such as BMP7 a TGFB1 antagonist, MMP8 a collagen breakdown inducer and follistatin, an inhibitor of the profibrotic factor myostatin. In conclusion, the addition of 1,25D to differentiated MMCs displays a decreased profibrotic signaling pathway and gene expression, leading to decrease in collagen deposition. This study highlights key mechanistic pathways through which vitamin D decreases fibrosis, and provides a rationale for studies to test vitamin D supplementation as a preventive and/or early treatment strategy for CVD and related fibrotic disorders.
Collapse
Affiliation(s)
- Jorge N Artaza
- Department of Internal Medicine, Charles Drew University of Medicine & Science, Los Angeles, California 90059, USA.
| | | |
Collapse
|
25
|
Abstract
Stem cell-based regenerative medicine holds great promise for repair of diseased tissue. Modern directions in the field of epigenetic research aimed to decipher the epigenetic signals that give stem cells their unique ability to self-renew and differentiate into different cell types. However, this research is only the tip of the iceberg when it comes to writing an 'epigenetic instruction manual' for the ramification of molecular details of cell commitment and differentiation. In this review, we discuss the impact of the epigenetic research on our understanding of stem cell biology.
Collapse
Affiliation(s)
- Victoria V Lunyak
- Buck Institute for Age Research, 8001 Redwood Blvd, Novato, CA 94945, USA.
| | | |
Collapse
|
26
|
Pluripotency Associated Genes Are Reactivated by Chromatin-Modifying Agents in Neurosphere Cells. Stem Cells 2008; 26:920-6. [DOI: 10.1634/stemcells.2007-0649] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
27
|
Dinger TC, Eckardt S, Choi SW, Camarero G, Kurosaka S, Hornich V, McLaughlin KJ, Müller AM. Androgenetic embryonic stem cells form neural progenitor cells in vivo and in vitro. Stem Cells 2008; 26:1474-83. [PMID: 18369101 DOI: 10.1634/stemcells.2007-0877] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Uniparental zygotes with two paternal (androgenetic [AG]) or two maternal (gynogenetic [GG]; parthenogenetic [PG]) genomes are not able to develop into viable offspring but can form blastocysts from which embryonic stem cells (ESCs) can be derived. Although some aspects of the in vitro and in vivo differentiation potential of PG and GG ESCs of several species have been studied, the developmental capacity of AG ESCs is much less clear. Here, we investigate the potential of murine AG ESCs to undergo neural differentiation. We observed that AG ESCs differentiate in vitro into pan-neural progenitor cells (pnPCs) that further give rise to cells that express neuronal- and astroglial-specific markers. Neural progeny of in vitro-differentiated AG ESCs exhibited fidelity of expression of six imprinted genes analyzed, with the exception of Ube3a. Bisulfite sequencing for two imprinting control regions suggested that pnPCs predominantly maintained their methylation pattern. Following blastocyst injection of AG and biparental (normal fertilized [N]) ESCs, we found widespread and evenly distributed contribution of ESC-derived cells in both AG and N chimeric early fetal brains. AG and N ESC-derived cells isolated from chimeric fetal brains by fluorescence-activated cell sorting exhibited similar neurosphere-initiating cell frequencies and neural multilineage differentiation potential. Our results indicate that AG ESC-derived neural progenitor/stem cells do not differ from N neural progenitor/stem cells in their self-renewal and neural multilineage differentiation potential. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Timo C Dinger
- Institut für Medizinische Strahlenkunde und Zellforschung, University of Würzburg, Versbacher Strasse 5, 97078 Würzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Boquest AC, Noer A, Collas P. Epigenetic programming of mesenchymal stem cells from human adipose tissue. ACTA ACUST UNITED AC 2007; 2:319-29. [PMID: 17848719 DOI: 10.1007/bf02698059] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/01/2023]
Abstract
Stromal stem cells identified in various adult mesenchymal tissues (commonly called mesenchymal stem cells [MSCs]) have in past years received more attention as a result of their potential interest as replacement cells in regenerative medicine. An abundant and easily accessible source of adult human MSCs are stem cells harvested from liposuction material. Similarly to bone marrow-derived MSCs, human adipose tissue-derived stem cells (ASCs) can give rise to a variety of cell types in vitro and in vivo; however, they have a propensity to differentiate into primarily mesodermal lineages. Even so, their capacity to differentiate into nonadipogenic mesodermal pathways seems to be restricted. Emerging DNA methylation profiles at adipogenic and nonadipogenic gene promoters in freshly isolated, cultured, or differentiated ASCs aim to provide an epigenetic explanation for this restrictive differentiation potential. A review of these studies indicates that human ASCs are epigenetically marked by mosaic hypomethylation of adipogenic promoters, whereas nonadipogenic lineage-specific promoters are hypermethylated. Surprisingly, in vitro differentiation toward various pathways maintains the overall methylation profiles of undifferentiated cells, raising the hypothesis that ASCs are at least epigenetically preprogrammed for adipogenesis. Novel attempts at reprogramming the epigenome of MSCs have been initiated to enhance the differentiation capacity of these cells.
Collapse
Affiliation(s)
- Andrew C Boquest
- Institute of Basic Medical Sciences, Department of Biochemistry, Faculty of Medicine, University of Oslo, Blindern, 0317 Oslo, Norway
| | | | | |
Collapse
|
29
|
Brulport M, Schormann W, Bauer A, Hermes M, Elsner C, Hammersen FJ, Beerheide W, Spitkovsky D, Härtig W, Nussler A, Horn LC, Edelmann J, Pelz-Ackermann O, Petersen J, Kamprad M, von Mach M, Lupp A, Zulewski H, Hengstler JG. Fate of extrahepatic human stem and precursor cells after transplantation into mouse livers. Hepatology 2007; 46:861-870. [PMID: 17668884 DOI: 10.1002/hep.21745] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
UNLABELLED In recent years, a large number of groups studied the fate of human stem cells in livers of immunodeficient animals. However, the interpretation of the results is quite controversial. We transplanted 4 different types of human extrahepatic precursor cells (derived from cord blood, monocytes, bone marrow, and pancreas) into livers of nonobese diabetic/severe combined immunodeficiency mice. Human hepatocytes were used as positive controls. Tracking of the transplanted human cells could be achieved by in situ hybridization with alu probes. Cells with alu-positive nuclei stained positive for human albumin and glycogen. Both markers were negative before transplantation. However, cells with alu-positive nuclei did not show a hepatocyte-like morphology and did not express cytochrome P450 3A4, and this suggests that these cells represent a mixed cell type possibly resulting from partial transdifferentiation. Using antibodies specific for human albumin, we also observed a second human albumin-positive cell type that could be clearly distinguished from the previously described cells by its hepatocyte-like morphology. Surprisingly, these cells had a mouse and not a human nucleus which is explained by transdifferentiation of human cells. Although it has not yet been formally proven, we suggest horizontal gene transfer as a likely mechanism, especially because we observed small fragments of human nuclei in mouse cells that originated from deteriorating transplanted cells. Qualitatively similar results were obtained with all 4 human precursor cell types through different routes of administration with and without the induction of liver damage. CONCLUSION We observed evidence not for transdifferentiation but instead for a complex situation including partial differentiation and possibly horizontal gene transfer.
Collapse
Affiliation(s)
- Marc Brulport
- Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Snykers S, Vinken M, Rogiers V, Vanhaecke T. Differential role of epigenetic modulators in malignant and normal stem cells: a novel tool in preclinical in vitro toxicology and clinical therapy. Arch Toxicol 2007; 81:533-44. [PMID: 17387455 DOI: 10.1007/s00204-007-0195-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Accepted: 02/22/2007] [Indexed: 02/06/2023]
Abstract
Adult stem cells are primitive cells that undergo asymmetric division, thereby giving rise to one clonogenic, self-renewing cell and one cell able to undergo multipotent differentiation. Disturbance of this controlled process by epigenetic alterations, including imbalance of histone acetylation/histone deacetylation and DNA methylation/demethylation, may result in uncontrolled growth, formation of self-renewing malignant stem cells and eventually cancer. In view of this notion, several epigenetic modulators, in particular those with histone deacetylase inhibiting activity, are currently being tested in phase I and II clinical trials for their promising chemotherapeutic properties in cancer therapy. As chromatin modulation is also involved in regulation of differentiation, normal development, embryonic and adult stem cell functions and maintenance of their plasticity during embryonic organogenesis, the question can be raised whether predestined cell fate can be modified through epigenetic interference. And if so, could this strategy enforce adult stem cells to differentiate into different types of functional cells? In particular, functional hepatocytes seem important for preclinical toxicity screening of candidate drugs. This paper reviews the potential use and relevance of epigenetic modifiers, including inhibitors of histone deacetylases and DNA methyltransferases (1) to change cell fate and 'trans'differentiate normal adult stem cells into hepatocyte-like cells and (2) to cure disorders, caused by uncontrolled growth of malignant stem cells.
Collapse
Affiliation(s)
- Sarah Snykers
- Department of Toxicology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | | | | | | |
Collapse
|
31
|
Abstract
Germ cell tumours of the brain and those that occur in the gonads are believed to share a common origin from germ cell progenitors. This 'germ cell theory' rests upon similar histopathology between these tumours in different locations and the belief that endogenous somatic cells of the brain could not give rise to the range of cell types seen in germ cell tumours. An alternative 'embryonic cell theory' has been proposed for some classes of cranial germ cell tumours, but this still relies on the misplacement of cells in the brain (in this case the earliest embryonic stem cells) during early embryonic development. Recent evidence has demonstrated that neural stem cells of the brain can also give rise to many of the cell types seen in germ cell tumours. These data suggest that endogenous progenitor cells of the brain are a plausible alternative origin for these tumours. This idea is of central importance for studies aiming to elucidate the mechanisms of tumour development. The application of modern molecular analyses to reveal how tumour cells have altered with respect to their cell of origin relies on the certain identification of the cell from which the particular tumour arose. If the identity of this cell is mistaken, then studies to elucidate the mechanisms by which the progenitor cell has been subverted from its normal behaviour will not yield useful information. In addition, it will prove impossible to generate an appropriate animal model in which to study the underlying causes of those tumours. This article makes the case that current assumptions of the origins of cranial germ cell tumours are unreliable. It reviews the evidence in favour of the 'germ cell theory' and argues in favour of a 'brain cell theory' in which endogenous neural progenitor cells of the brain are the likely origin for these tumours. Thus, the case is made that cranial germ cell tumours, like other brain tumours, arise by the transformation of progenitor cells normally resident in the brain.
Collapse
Affiliation(s)
- P J Scotting
- Children's Brain Tumour Research Centre, Institute of Genetics, University of Nottingham, Nottingham, UK.
| |
Collapse
|
32
|
Bonifer C, Lefevre P, Tagoh H. The Regulation of Chromatin and DNA-Methylation Patterns in Blood Cell Development. Curr Top Microbiol Immunol 2006; 310:1-12. [PMID: 16909903 DOI: 10.1007/3-540-31181-5_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
All developmental processes in metazoans require the establishment of different genetic programs to generate functionally specialised cells. Differential gene expression is also the basis for the alterations in the developmental potential of differentiating cells. However, the molecular details concerning how this is achieved are still poorly understood. The haematopoietic system has for many years served as an excellent model system to studyhow developmental processes are regulated at the epigenetic level. In this article we will summarise recent results from others and from our own laboratory that have yielded profound insights into the general principles of how cell-fate decisions are regulated in the cell nucleus. We summarise (1) how the interplay of sequence-specific transcription factors and chromatin components is responsible for the cell type and cell stage-specific activation of specific genes and (2) how these findings impact on current concepts of epigenetic regulation of developmental processes.
Collapse
Affiliation(s)
- C Bonifer
- Division of Experimental Haematology, Institute for Molecular Medicine, Epidemiology and Cancer Research, University of Leeds, St James's University Hospital, UK.
| | | | | |
Collapse
|
33
|
Bryder D, Rossi DJ, Weissman IL. Hematopoietic stem cells: the paradigmatic tissue-specific stem cell. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:338-46. [PMID: 16877336 PMCID: PMC1698791 DOI: 10.2353/ajpath.2006.060312] [Citation(s) in RCA: 439] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The recent prospective isolation of a wide variety of somatically derived stem cells has affirmed the notion that homeostatic maintenance of most tissues and organs is mediated by tissue-specific stem and progenitor cells and fueled enthusiasm for the use of such cells in strategies aimed at repairing or replacing damaged, diseased, or genetically deficient tissues and organs. Hematopoietic stem cells (HSCs) are arguably the most well-characterized tissue-specific stem cell, with decades of basic research and clinical application providing not only a profound understanding of the principles of stem cell biology, but also of its potential pitfalls. It is our belief that emerging stem cell fields can benefit greatly from an understanding of the lessons learned from the study of HSCs. In this review we discuss some general concepts regarding stem cell biology learned from the study of HSCs with a highlight on recent work pertaining to emerging topics of interest for stem cell biology.
Collapse
Affiliation(s)
- David Bryder
- Stanford University School of Medicine, B257 Beckman Center, Stanford, CA 94305-5323, USA
| | | | | |
Collapse
|
34
|
Lotem J, Sachs L. Epigenetics and the plasticity of differentiation in normal and cancer stem cells. Oncogene 2006; 25:7663-72. [PMID: 16847453 DOI: 10.1038/sj.onc.1209816] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Embryonic stem cells are characterized by their differentiation to all cell types during embryogenesis. In adult life, different tissues also have somatic stem cells, called adult stem cells, which in specific niches can undergo multipotent differentiation. The use of these adult stem cells has considerable therapeutic potential for the regeneration of damaged tissues. In both embryonic and adult stem cells, differentiation is controlled by epigenetic mechanisms, and the plasticity of differentiation in these cells is associated with transcription accessibility for genes expressed in different normal tissues. Abnormalities in genetic and/or epigenetic controls can lead to development of cancer, which is maintained by self-renewing cancer stem cells. Although the genetic abnormalities produce defects in growth and differentiation in cancer stem cells, these cells have not always lost the ability to undergo differentiation through epigenetic changes that by-pass the genomic abnormalities, thus creating the basis for differentiation therapy. Like normal stem cells, cancer stem cells can show plasticity for differentiation. This plasticity of cancer stem cells is also associated with transcription accessibility for genes that are normally expressed in different tissues, including tissues other than those from which the cancers originated. This broad transcription accessibility can also contribute to the behavior of cancer cells by overexpressing genes that promote cell viability, growth and metastasis.
Collapse
Affiliation(s)
- J Lotem
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
35
|
Kondo T. Epigenetic alchemy for cell fate conversion. Curr Opin Genet Dev 2006; 16:502-7. [PMID: 16844365 DOI: 10.1016/j.gde.2006.07.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Accepted: 07/05/2006] [Indexed: 12/15/2022]
Abstract
Recent progress in neural stem cell research shows that a number of extrinsic factors and intracellular mechanisms, including epigenetic modifications, are involved in the self-renewal of neural stem cells and in neuronal and glial differentiation. Remarkably, there is increasing evidence that the remodeling of chromatin structure and the alteration of epigenetic marks, including histone methylation and acetylation and DNA methylation, can cause committed cells to convert from one fate to another, and such converted cells are functional when transplanted in vivo. Thus, epigenetic research might generate the alchemy required to convert any non-neural stem cells into functional neural stem cells, which are few and difficult to extract from the adult central nervous system.
Collapse
Affiliation(s)
- Toru Kondo
- Laboratory for Cell Lineage Modulation, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| |
Collapse
|