Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Invest New Drugs ; 41(3): 371-375, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37162706

RESUMEN

Glioblastomas (GBM), also known as glioblastoma multiforme, are the most aggressive type of brain cancer. Currently, there is no effective treatment for GBM, highlighting the pressing need for new therapeutic strategies. In a recent study, we demonstrated that specific combinations of epigenetic modifiers significantly affect the metabolism and proliferation rate of the two most aggressive GBM cell lines, D54 and U-87. Importantly, these combinations exhibited minimal effects on the growth of normal stem cells. In this study, we extended our investigation to include a patient-derived GBM stem cell line. Our results showed that the combinations of modulators of histone and DNA covalent modifying enzymes that synergistically suppress D54 and U87 cell line growth also impair the viability of the patient-derived GBM stem cell line. These findings suggest that epigenetic modifiers alone or in specific combinations exhibit a cytotoxic effect on established and low-passage patient-derived GBM cell lines, and thus could be a promising therapeutic approach for this type of brain cancer.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Proliferación Celular , Línea Celular , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Epigénesis Genética , Células Madre/metabolismo , Línea Celular Tumoral
2.
Mol Cell Biochem ; 477(10): 2309-2317, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35503191

RESUMEN

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.


Asunto(s)
Reprogramación Celular , Histonas , Adenosina Trifosfato/metabolismo , Animales , Cromatina , Metilación de ADN , Epigénesis Genética , Histonas/metabolismo , Mamíferos/genética , Mamíferos/metabolismo , Transducción de Señal
3.
Tumour Biol ; 36(11): 9067-72, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26084611

RESUMEN

The discoveries of recent decade showed that all critical changes in cancer cells, such as silencing of tumor-suppressor genes and activation of oncogenes, are caused not only by genetic but also by epigenetic mechanisms. Although epigenetic changes are somatically heritable, in contrast to genetic changes, they are potentially reversible, making them good targets for therapeutic intervention. Covalent modifications of chromatin such as methylation and acetylation of histones and methylation of DNA are the important components of epigenetic machinery. In this study, we investigated the effect of different modulators of DNA and histone covalent-modifying enzymes on the proliferation and viability of normal adult stem cells, such as human bone marrow mesenchymal stem cells (hMSCs), and on malignant tumor cells, such as glioblastoma (GB) D54 cells. Results demonstrated that specific combinations of histone methyltransferases and deacetylases inhibitors significantly attenuated D54 cells viability but having only a small effect on hMSCs growth. Taken together, these studies suggest that specific combinations of histone covalent modifiers could be an effective treatment option for the most aggressive type of primary brain tumors such as glioblastoma multiforme.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Epigénesis Genética , Glioblastoma/genética , Inhibidores de Histona Desacetilasas/administración & dosificación , Histonas/metabolismo , Células Madre Adultas/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromatina/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Histona Desacetilasas/metabolismo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Histonas/antagonistas & inhibidores , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos
4.
Cells Dev ; : 203928, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38768658

RESUMEN

Inheritance of acquired characteristics is the once widely accepted idea that multiple modifications acquired by an organism during its life, can be inherited by the offspring. This belief is at least as old as Hippocrates and became popular in early 19th century, leading Lamarck to suggest his theory of evolution. Charles Darwin, along with other thinkers of the time attempted to explain the mechanism of acquired traits' inheritance by proposing the theory of pangenesis. While later this and similar theories were rejected because of the lack of hard evidence, the studies aimed at revealing the mechanism by which somatic information can be passed to germ cells have continued up to the present. In this paper, we present a new theory and provide supporting literature to explain this phenomenon. We hypothesize existence of pluripotent adult stem cells that can serve as collectors and carriers of new epigenetic traits by entering different developmentally active organ/tissue compartments through blood circulation and acquiring new epigenetic marks though cycles of differentiation/dedifferentiation or transdifferentiation. During gametogenesis, these epigenetically modified cells are attracted by gonads, transdifferentiate into germ cells, and pass the acquired epigenetic modifications collected from the entire body's somatic cells to the offspring.

5.
J Cell Mol Med ; 16(11): 2708-14, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22681532

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Neuronas Dopaminérgicas/fisiología , Epigénesis Genética , Células Madre Mesenquimatosas/fisiología , Biomarcadores/metabolismo , Células de la Médula Ósea/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipoxia de la Célula , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Neuronas Dopaminérgicas/citología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo
6.
Med Oncol ; 39(5): 86, 2022 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-35478054

RESUMEN

Discoveries made over the last decade have shown that critical changes in cancer cells, such as activation of oncogenes and silencing of tumor suppressor genes are caused not only by genetic but also by epigenetic mechanisms. While epigenetic alterations are somatically heritable, in contrast to genetic changes, they are potentially reversible, making them perfect targets for therapeutic intervention. Covalent modifications of chromatin, such as methylation of DNA and acetylation and methylation of histones, are important components of epigenetic machinery. Multiple recent studies have shown that epigenetic modifiers are candidates for potent new drugs in multiple cancers' therapies, including gliomas, and several clinical trials are ongoing. However, as with other chemotherapeutic drugs, toxicity is one of the main concerns with some of the potent epigenetic drugs. Synergistic combinations of these agents are one approach to overcoming toxicity issues while enhancing efficacy. In this study, we demonstrated that while individually BIX01294, an inhibitor of histone methyltransferase G9a, DZNep, an inhibitor of lysine methyltransferase EZH2, and Trichostatin A (TSA), an inhibitor of histone deacetylase at their low concentrations showed a moderate effect on the viability of U87 glioblastoma cells, in combinations they exhibited a synergistic effect. Importantly, these combinations exhibited minimal effect on adipose mesenchymal stem cells (AD-MSCs) growth. Thus, unique combinations and concentrations of epigenetic modifiers, that synergistically attenuated the U87 glioblastoma cells while exhibiting minor or moderate effects on normal stem cell growth, have been discovered.


Asunto(s)
Glioblastoma , Células Madre Mesenquimatosas , Línea Celular , Metilación de ADN , Epigénesis Genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Humanos
7.
Assay Drug Dev Technol ; 20(2): 75-82, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35196113

RESUMEN

Migration is a key property of live cells and critical for normal development, immune response, and disease processes such as cancer metastasis and inflammation. Methods to examine cell migration are especially useful and important for a wide range of biomedical research such as cancer biology, immunology, vascular biology, cell biology, and developmental biology. In vitro assays are excellent approaches to extrapolate to in vivo situations and study live cells behavior. The aim of this article is to discuss the existing methods for transwell migration/invasion studies, the problems associated with this assay, and proposed modifications to this methodological approach that makes it simple to perform and improve the assay accuracy. Results of our studies demonstrated that the count of cells that had grown on top of the membrane is important to accurately evaluate the percentage of migrated/invaded cells. The results also showed that the transparent transwell insert with 4',6-diamidino-2-phenylindole (DAPI) stained cells is the best approach to ease the analysis of cell numbers on top of the membranes. In addition, the overlay of bright light (representing membrane pores) and DAPI images can further improve the accuracy of cell count. All these modifications in combination simplify the assay performance and improve the accuracy of the transwell migration assay method.


Asunto(s)
Movimiento Celular , Recuento de Células , Línea Celular Tumoral , Movimiento Celular/fisiología
8.
Cytotherapy ; 13(1): 61-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20735167

RESUMEN

BACKGROUND AIMS: Traumatic injuries of the central nervous system cause damage and degeneration of specific cell populations with subsequent functional loss. Cell transplantation is a strategy to treat such injuries by replacing lost or damaged cell populations. Many kinds of cells are considered candidates for intraspinal transplantation. Human neural precursor cells (hNPC) derived from post-mortem fetal tissue are easy to isolate and expand, and are capable of producing large numbers of neuronal and glial cells. After transplantation into the nervous system, hNPC produce mature neural phenotypes and permit functional improvement in some models of neurodegenerative disease. In this study, we aimed to elucidate the therapeutic effect of different neuronal and glial progenitor populations of hNPC on locomotor and sensory functions of spinal cord-injured (SCI) rats. METHODS: Different populations of progenitor cells were obtained from hNPC by cell sorting and neural induction, resulting in cell cultures that were NCAM(+) A2B5(+), NCAM(+) A2B5(-) or A2B5(+) NG2(+). These different cell populations were then tested for efficacy in repair of the injured spinal cord by transplantation into rats with SCI. RESULTS: The A2B5(+) NG2(+) population of hNPC significantly improved locomotor and sensory (hindlimb) functional recovery of SCI rats. Importantly, no abnormal pain responses were observed in the forelimbs following transplantation. CONCLUSIONS: This treatment approach can improve functional recovery after SCI without causing allodynia. Further studies will be conducted to investigate the ability of A2B5(+) NG2(+) cells to survive, differentiate and integrate in the injured spinal cord.


Asunto(s)
Hiperalgesia/complicaciones , Células-Madre Neurales/citología , Neuroglía/trasplante , Recuperación de la Función/fisiología , Sensación/fisiología , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre , Animales , Conducta Animal , Biomarcadores/metabolismo , Proliferación Celular , Supervivencia Celular , Femenino , Miembro Anterior/fisiopatología , Miembro Posterior/fisiopatología , Humanos , Hiperalgesia/fisiopatología , Actividad Motora/fisiología , Células-Madre Neurales/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/fisiopatología
9.
J Neurosci Methods ; 150(1): 41-6, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16087243

RESUMEN

Several recent studies have shown that highly undifferentiated neural stem cells (NSCs) grafted into the intact or injured adult spinal cord of animal either remain undifferentiated or show fate restriction to a astrocytic lineage. This indicates that functionally diverse roles expected of cellular replacement cannot be performed by the transplantation of highly immature precursors; rather, more differentiated or appropriate mixtures of more restricted neural precursors may be important in replacement strategies. In this study, we investigated the ability of lineage-restricted neural progenitors derived from adult mouse periventricular subependymal zone (SEZ) to integrate and differentiate into the chronically injured adult spinal cord. To this end, NSCs were grown as adherent cultures followed by expansion in non-adhesive dishes. This allowed us to grow NSCs as colonies of restricted neural precursors, illustrated by NCAM, nestin, Sox-2, A2B5, and GFAP immunostaining. The mixture of lineage-restricted precursors was directly implanted into the chronically injured spinal cord of immunosuppressed cats. The fate of the cells was traced with GFP fluorescence and immunocytochemistry for neural markers such as beta-III-tubulin, GFAP, and Ng2. After four weeks, transplanted cells survived, giving rise to neurons and in addition to cells with an astrocytic phenotype. We conclude that a mixture of more restricted neural precursors may be better suited than highly immature NSCs for neural replacement strategies after central nervous system (CNS) injuries.


Asunto(s)
Supervivencia de Injerto , Neuronas/citología , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre , Animales , Astrocitos/citología , Gatos , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Femenino , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Transgénicos , Oligodendroglía/citología , Células Madre/citología
10.
Int J Biochem Cell Biol ; 64: 190-4, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25936755

RESUMEN

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.


Asunto(s)
Epigénesis Genética , Células Madre Mesenquimatosas/fisiología , Adipocitos/metabolismo , Diferenciación Celular , Células Cultivadas , Humanos , Fenotipo
11.
J Tissue Eng Regen Med ; 8(5): 407-13, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-22674835

RESUMEN

Mesenchymal stem cells (MSCs) in their immature state express a variety of genes of the three germ layers at relatively low or moderate levels that might explain their phenomenal plasticity. Numerous recent studies have demonstrated that under the appropriate conditions in vitro and in vivo the expression of different sets of these genes can be upregulated, turning MSCs into variety of cell lineages of mesodermal, ectodermal and endodermal origin. While transdifferentiation of MSCs is still controversial, these unique properties make MSCs an ideal autologous source of easily reprogrammable cells. Recently, using the approach of cell reprogramming by biological active compounds that interfere with chromatin structure and function, as well as with specific signalling pathways that promote neural fate commitment, we have been able to generate neural-like cells from human bone marrow (BM)-derived MSCs (hMSCs). However, the efficiency of neural transformation of hMSCs induced by this approach gradually declined with passaging. To elucidate the mechanisms that underlie the higher plasticity of early-passage hMSCs, comparative analysis of the expression levels of several pluripotent and neural genes was conducted for early- and late-passage hMSCs. The results demonstrated that early-passage hMSCs expressed the majority of these genes at low and moderate levels that gradually declined at late passages. Neural induction further increased the expression of some of these genes in hMSCs, accompanied by morphological changes into neural-like cells. We concluded that low and moderate expression of several pluripotent and neural genes in early-passage hMSCs could explain their higher plasticity and pliability for neural induction.


Asunto(s)
Células de la Médula Ósea/citología , Ensamble y Desensamble de Cromatina , Células Madre Mesenquimatosas/citología , Plasticidad Neuronal , Proteínas Nucleares/metabolismo , Western Blotting , Forma de la Célula , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Transl Res ; 162(5): 317-23, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24001408

RESUMEN

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.


Asunto(s)
AMP Cíclico/metabolismo , Dopamina/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Medios de Cultivo , Factor 8 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Hedgehog/farmacología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Smad/metabolismo
13.
Int J Biochem Cell Biol ; 45(8): 1633-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23665234

RESUMEN

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.


Asunto(s)
Reprogramación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/efectos de los fármacos , AMP Cíclico/metabolismo , Células Madre Mesenquimatosas/citología , Neuronas/citología , Proteínas Smad/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Western Blotting , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Activación del Canal Iónico/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/enzimología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Sodio/metabolismo , Sinapsis/metabolismo
14.
Neurorehabil Neural Repair ; 25(9): 873-80, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21844281

RESUMEN

BACKGROUND: Stem cell-based therapy for repair and replacement of lost neural cells is a promising treatment for central nervous system (CNS) diseases. Bone marrow (BM)-derived mesenchymal stem cells (MSCs) can differentiate into neural phenotypes and be isolated and expanded for autotransplantation with no risk of rejection. OBJECTIVE: The authors examined whether transplanted neurally induced human MSCs (NI hMSCs), developed by a new procedure, can survive, differentiate, and promote tissue protection and functional recovery in injured spinal cord (ISC) rats. METHODS: Neural induction was achieved by exposing cells simultaneously to inhibitors of DNA methylation, histone deacetylation, and pharmacological agents that increased cAMP levels. Three groups of adult female Sprague-Dawley rats were injected immediately rostral and caudal to the midline lesion with phosphate-buffered saline, MSCs, or NI hMSCs, 1 week after a spinal cord impact injury at T-8. Functional outcome was measured using the Basso Beattie Bresnahan (BBB) locomotor rating scale and thermal sensitivity test on a weekly basis up to 12 weeks postinjury. Graft integration and anatomy of spinal cord was assessed by stereological, histochemical, and immunohistochemical techniques. RESULTS: The transplanted NI hMSCs survived, differentiated, and significantly improved locomotor recovery of ISC rats. Transplantation also reduced the volume of lesion cavity and white matter loss. CONCLUSION: This method of hMSC modification may provide an alternative source of autologous adult stem cells for CNS repair.


Asunto(s)
Trasplante de Médula Ósea/patología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Neurogénesis/fisiología , Parálisis/cirugía , Traumatismos de la Médula Espinal/cirugía , Animales , Trasplante de Médula Ósea/métodos , Femenino , Humanos , Células Madre Mesenquimatosas/patología , Parálisis/patología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/patología , Trasplante Heterólogo/patología , Resultado del Tratamiento
15.
Cell Reprogram ; 13(5): 385-90, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21718099

RESUMEN

Recent studies suggest that cellular therapies that utilize mesenchymal stem cells (MSCs), especially ones that have been neurally induced (NI), may provide a functional benefit in a wide range of neurological disorders. Recently, we developed a new method for the efficient generation of neural cells from human bone marrow (BM)-derived MSCs (hMSC). Neural induction was achieved by exposing cells simultaneously to chromatin-modifying agents and neural-inducing factors. When transplanted into injured spinal cords, these NI-hMSCs survived, differentiated, promoted tissue preservation, and significantly improved locomotor recovery of injured animals. In the current study, we sought to determine whether this methodological approach would be equally effective in generating neural-like cells from feline BM-derived MSCs (fMSC). Our long-term goal is to develop an autologous source of neural stem cells that can be used in cellular replacement therapies in large animal (feline) models of neurological disorders. Our results showed that fMSCs exhibited a neural morphology after 48-72 h of neural induction. Immunocytochemistry, ELISA, Western blot, and real-time RT-PCR studies revealed a higher level of expression of several pluripotent and neural genes in NI-fMSCs, the majority of which were expressed in untreated fMSCs at relatively low levels. We concluded that the expression of pluripotency- and neural-associated genes in unmodified fMSCs make them more pliable for reprogramming into a neural fate by manipulation with chromatin modifying agents and neural inducing factors.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Células de la Médula Ósea/metabolismo , Cromatina/metabolismo , Inhibidores Enzimáticos/farmacología , Células Madre Mesenquimatosas/metabolismo , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Células de la Médula Ósea/citología , Gatos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Traumatismos de la Médula Espinal/terapia , Trasplante Autólogo
16.
Regen Med ; 5(6): 891-900, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21082889

RESUMEN

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.


Asunto(s)
Células Madre Adultas/citología , Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/citología , Neuronas/citología , Adulto , Células Madre Adultas/metabolismo , Western Blotting , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Forma de la Célula , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/metabolismo , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Coloración y Etiquetado
17.
Restor Neurol Neurosci ; 28(6): 761-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21209491

RESUMEN

PURPOSE: We recently developed a new method for efficient generation of neural-like cells from mice bone marrow (BM)-derived mesenchymal stem cells (MSC) by exposing MSCs to epigenetic modifiers and a neural stem cell environment. These neurally induced MSCs (NI-MSCs) differentiate into neuronal- and glial-like cells in vitro, release neurotrophic factors NGF and BDNF, survive and integrate after transplantation in intact spinal cord. The aim of this study was to determine whether transplanted NI-MSCs survive, differentiate, and integrate in injured spinal cord (ISC) rats and promote functional recovery. METHODS: Twenty rats, half grafted with MSCs and half with NI-MSCs, were used for survival and differentiation studies. Results were analyzed using triple-labeled immunohistochemistry. For motor function studies the 3 group of adult female Sprague Dawley rats received PBS (vehicle), MSCs, or NI-MSCs, respectively. Functional outcome was measured using the BBB scale. RESULTS: Results demonstrated gradual improvement of locomotor function in NI-MSC-transplanted rats in comparison to vehicle and non-modified MSC-transplanted animals, with statistically significant differences at 7, 14, and 21 days post transplantation. Immunocytochemical studies revealed poor survival of NI-MSCs within the ISC as early as 3 weeks after transplantation. CONCLUSIONS: Thus, there is a correlation between the degree of surviving NI-MSCs and extent of functional recovery.


Asunto(s)
Diferenciación Celular/fisiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células-Madre Neurales/citología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/terapia , Análisis de Varianza , Animales , Supervivencia Celular/fisiología , Femenino , Inmunohistoquímica , Actividad Motora/fisiología , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/fisiopatología
18.
Stem Cells Dev ; 17(6): 1123-30, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18484898

RESUMEN

Mesenchymal stem cell (MSC)-mediated tissue regeneration is a promising strategy to treat several neurodegenerative diseases and traumatic injuries of the central nervous system. Bone marrow MSCs have great potential as therapeutic agents, since they are easy to isolate and expand and are capable of producing various cell types, including neural cells. Recently we developed a highly efficient methodology to produce neural stem-like and neural precursor-like cells from mice bone marrow-derived MSCs that eventually differentiate into neuronal- and glial-like cells in vitro. The aim of this study is to further elucidate neural expression profile of neurally induced mesenchymal stem cells (NI-MSCs) and their ability to retain neural differentiation potential when grafted into the intact spinal cord of rats. To this end, we further characterized in vitro and in vivo properties of NI-MSCs by immunocytochemistry, Western blot, ELISA, and immunohistochemistry. Immunocytochemical data demonstrated that NI-MSCs express several mature neural markers such as B3T, GFAP MAP-2, NF-200, and NeuN, which were confirmed through Western blot. ELISA data showed that NI-MSCs release nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF). In vivo studies demonstrated that grafted NI-MSCs survived after transplantation into intact spinal cord and produced cells that expressed neural markers. All these data suggest that neurally modified MSCs, induced by recently developed methodology, could be a potential source of cells to replace damaged neurons and glia in injured spinal cord, and/or to promote cell survival and axonal growth of host tissue.


Asunto(s)
Diferenciación Celular/fisiología , Epigénesis Genética , Células Madre Mesenquimatosas/citología , Tejido Nervioso/citología , Animales , Antígenos de Diferenciación/metabolismo , Células de la Médula Ósea , Supervivencia Celular/fisiología , Células Cultivadas , Supervivencia de Injerto/fisiología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/terapia , Ratas , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/métodos
19.
J Cell Biochem ; 100(2): 362-71, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16924670

RESUMEN

Understanding mechanisms that govern cell fate decisions will lead to developing techniques for induction of adult stem cell differentiation to desired cell outcomes and, thus, production of an autologos source of cells for regenerative medicine. Recently, we demonstrated that stem cells derived from adult central nervous system or bone marrow grown with other cell lineages or with more undifferentiated cells sometimes take on those characteristics. This indicates that manipulating extracellular factors may be sufficient to alter some developmental restrictions regulated by the epigenetic system. In this study, using pharmacological agents that interfere with the main components of the epigenetic program such as DNA methylation and histone deacetylation, we induce high-level expression of embryonic and neural stem cell (NSC) marker Sox2 in bone marrow-derived mesenchymal stem cells (MSCs). Exposure of these modified cells to a neural environment via juxtacrine and paracrine interactions promote efficient generation of neural stem-like cells as well as cells with neuronal and glial characteristics. We concluded that the manipulation strategy used in this study can be a useful method for efficient production of NSC-like cells from MSCs.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Epigénesis Genética , Células Madre Mesenquimatosas/citología , Neuronas/citología , Animales , Células de la Médula Ósea/metabolismo , Proliferación Celular , Forma de la Célula , Células Cultivadas , Medios de Cultivo Condicionados , Proteínas de Unión al ADN/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Neuronas/metabolismo , Factores de Transcripción SOXB1 , Transactivadores/metabolismo
20.
Exp Neurol ; 201(2): 335-48, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16839548

RESUMEN

Transplantation of neural stem cells (NSCs) in the injured spinal cord has been shown to improve functional outcome; however, recent evidence has demonstrated forelimb allodynia following transplantation of embryonic NSCs. The aim of this study was to investigate whether transplantation of murine C17.2 NSCs alone or transfected with glial-derived neurotrophic factor (C17.2/GDNF) would induce allodynia in transplanted spinal cord-injured animals. One week after a T8-level spinal cord injury (SCI), C17.2, C17.2/GDNF or normal saline was injected at the injury site. Locomotor function and sensory recovery to thermal and mechanical stimuli were then measured. Spinal cords were processed immunohistochemically at the injury/transplantation site for characterization of NSC survival and differentiation; and at the cervicothoracic level for calcitonin gene-related peptide (CGRP), a neuropeptide expressed in dorsal horn nocioceptive neurons, and growth-associated protein-43 (GAP43), a marker of neuronal sprouting. Locomotor function was not significantly improved following NSC transplantation at any time (P >0.05). Significant forelimb thermal and mechanical allodynia were observed following transplantation with both NSC populations (P <0.05). The C17.2 and C17.2/GDNF NSCs survived and differentiated into a predominately astrocytic population. Calcitonin gene-related peptide and GAP43 immunoreactivity significantly increased and co-localized in cervicothoracic dorsal horn laminae I-III following C17.2 and C17.2/GDNF transplantation. This study demonstrated that murine C17.2 NSCs differentiated primarily into astrocytes when transplanted into the injured spinal cord, and resulted in thermal and mechanical forelimb allodynia. Sprouting of nocioceptive afferents occurred rostral to the injury/transplantation site only in allodynic animals, suggesting a principal role in this aberrant pain state. Further, a difference in the degree of allodynia was noted between C17.2- and C17.2/GDNF transplant-treated groups; this difference correlated with the level of CGRP/GAP43 immunoreactivity and sprouting observed in the cervicothoracic dorsal horns. Both allodynia- and CGRP/GAP43-positive afferent sprouting were less in the C17.2/GDNF group compared to the C17.2 group, suggesting a possible protective or analgesic effect of GDNF on post-injury neuropathic pain.


Asunto(s)
Neuronas/citología , Dolor/fisiopatología , Traumatismos de la Médula Espinal/cirugía , Trasplante de Células Madre/métodos , Animales , Axones/metabolismo , Conducta Animal/fisiología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Supervivencia Celular/fisiología , Femenino , Miembro Anterior/fisiopatología , Proteína GAP-43/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Miembro Posterior/fisiopatología , Calor/efectos adversos , Inmunohistoquímica , Ratones , Actividad Motora/fisiología , Neuronas/metabolismo , Dolor/etiología , Células del Asta Posterior/citología , Células del Asta Posterior/metabolismo , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/fisiopatología , Trasplante de Células Madre/efectos adversos , Estrés Mecánico , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA