Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Biomed Sci ; 18: 54, 2011 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-21824437

RESUMEN

BACKGROUND: Spinocerebellar ataxia (SCA) refers to a disease entity in which polyglutamine aggregates are over-produced in Purkinje cells (PCs) of the cerebellum as well as other neurons in the central nervous system, and the formation of intracellular polyglutamine aggregates result in the loss of neurons as well as deterioration of motor functions. So far there is no effective neuroprotective treatment for this debilitating disease although numerous efforts have been made. Mesenchymal stem cells (MSCs) possess multi-lineage differentiation potentials as well as immuno-modulatory properties, and are theoretically good candidates for SCA treatment. The purpose of this study is to investigate whether transplantation of human MSCs (hMSCs) can rescue cerebellar PCs and ameliorate motor function deterioration in SCA in a pre-clinical animal model. METHOD: Transgenic mice bearing poly-glutamine mutation in ataxin-2 gene (C57BL/6J SCA2 transgenic mice) were serially transplanted with hMSCs intravenously or intracranially before and after the onset of motor function loss. Motor function of mice was evaluated by an accelerating protocol of rotarod test every 8 weeks. Immunohistochemical stain of whole brain sections was adopted to demonstrate the neuroprotective effect of hMSC transplantation on cerebellar PCs and engraftment of hMSCs into mice brain. RESULTS: Intravenous transplantation of hMSCs effectively improved rotarod performance of SCA2 transgenic mice and delayed the onset of motor function deterioration; while intracranial transplantation failed to achieve such neuroprotective effect. Immunohistochemistry revealed that intravenous transplantation was more effective in the preservation of the survival of cerebellar PCs and engraftment of hMSCs than intracranial injection, which was compatible to rotarod performance of transplanted mice. CONCLUSION: Intravenous transplantation of hMSCs can indeed delay the onset as well as improve the motor function of SCA2 transgenic mice. The results of this preclinical study strongly support further exploration of the feasibility to transplant hMSCs for SCA patients.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Actividad Motora/fisiología , Células de Purkinje/fisiología , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/terapia , Análisis de Varianza , Animales , Ataxinas , Compuestos Férricos , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Nanopartículas del Metal , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Péptidos/genética , Células de Purkinje/patología , Prueba de Desempeño de Rotación con Aceleración Constante
2.
Cell Transplant ; 21(5): 997-1009, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22004871

RESUMEN

Depletion of pancreatic ß-cells results in insulin insufficiency and diabetes mellitus (DM). Single transplantation of mesenchymal stem cells exhibits short-term effects in some preclinical studies. Here, we further investigated the long-term therapeutic effects of multiple intravenous MSC transplantations. In this study, multiple human MSC transplantations (4.2 × 10(7) cells/kg each time) were performed intravenously at 2-week intervals into streptozocin (STZ)-induced diabetic mice for 6 months. Blood sugar, insulin, renal function, cholesterol, and triglyceride levels were monitored. We demonstrated that compared to single intravenous transplantation, which only transiently decreased hyperglycemia, multiple MSC transplantations effectively restored blood glucose homeostasis. Systemic oxidative stress levels were reduced from the seventh week of treatment. From the 11th week, production of human insulin was markedly increased. When MSC transplantation was skipped after blood sugar level returned to normal at the end of 15th week, a sharp rebound of blood sugar occurred, and was then controlled by subsequent transplantations. At the end of 6 months, histopathology examination revealed MSCs specifically engrafted into liver tissues of the recipients. Fifty-one percent of human cells in the recipient liver coexpressed human insulin, especially those surrounding the central veins. Taken together, intravenous MSC delivery was safe and effective for blood glucose stabilization in this preclinical DM model. Multiple transplantations were essential to restore and maintain glucose homeostasis through decreasing systemic oxidative stress in the early stage and insulin production in the late stage. Liver engraftment and differentiation into insulin-producing cells account for the long-term therapeutic effects of MSCs.


Asunto(s)
Glucemia/análisis , Diabetes Mellitus Experimental/terapia , Células Secretoras de Insulina/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Diferenciación Celular , Colesterol/sangre , Humanos , Inyecciones Intravenosas , Insulina/sangre , Hígado/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo , Triglicéridos/sangre
3.
Cell Transplant ; 20(8): 1209-20, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21176396

RESUMEN

Mesenchymal stem cells (MSCs) are of great therapeutic potentials due to their multilineage differentiation capabilities. Before transplantation, in vitro culture expansion of MSCs is necessary to get desired cell number. We observed that cell contact accelerated replicative senescence during such process. To confirm the finding as well as to investigate the underlying mechanisms, we cultured both human bone marrow- and umbilical cord blood-derived MSCs under noncontact culture (subculture performed at 60-70% of confluence), or contact culture (cell passage performed at 100% of confluence). It was found that MSCs reached cellular senescence earlier in contact culture, and the doubling time was significantly prolonged. Marked increase of senescence-associated ß-galactosidase-positive staining was also observed as a result of cell contact. Cell cycle analysis revealed increased frequency of cell cycle arrest after contact culture. It was noted, however, that the telomere length was not altered during contact-induced acceleration of senescence. Moreover, cell cycle checkpoint regulator P53 expression was not affected by cell contact. Marked increase in intracellular reactive oxygen species (ROS) and a concomitant decrease in the activities of antioxidative enzymes were also observed during contact-induced senescence. Importantly, increased p16(INK4a) following Ras upregulation was found after contact culture. Taken together, cell contact induced accelerated senescence of MSCs, which is telomere shortening and p53 independent. ROS accumulation due to defective ROS clearance function together with Ras and p16(INK4a) upregulation play an important role in contact-induced senescence of MSCs. Overconfluence should therefore be avoided during in vitro culture expansion of MSCs in order to maintain their qualities for clinical application purposes. The contact-induced senescence model reported in this study will serve as a useful model system that allows further study of the molecular mechanisms of senescence in MSCs.


Asunto(s)
Senescencia Celular , Células Madre Mesenquimatosas/citología , Estrés Oxidativo , Acortamiento del Telómero , Proteínas ras/metabolismo , Adenosina Trifosfato/biosíntesis , Adolescente , Adulto , Antioxidantes/metabolismo , Células de la Médula Ósea/citología , Adhesión Celular , Puntos de Control del Ciclo Celular , Células Cultivadas , Conexina 43/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Fase G1 , Humanos , Espacio Intracelular/metabolismo , Células Madre Mesenquimatosas/enzimología , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Fase de Descanso del Ciclo Celular , Factores de Tiempo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Adulto Joven
4.
Tissue Eng Part A ; 17(1-2): 255-66, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20726817

RESUMEN

Loss of corneal epithelial cells results in visual problems. Stem cells isolated from the limbal area of the ocular surface are able to replenish lost corneal epithelial cells. However, destruction of the healthy limbus tissue is inevitable. Theoretically, orbital fat should be an excellent source to isolate stem cells for regenerating ocular tissues as the orbital connective tissues share the same embryonic origin with the ocular proper in early organogenesis. The aim of this study is to isolate stem cells from the human orbital fat and to explore their differentiation potentials into epithelial cells. It was found that spindle-shaped, fibroblast-like cells with extensive proliferation potentials could be isolated from orbital fat tissues. These orbital fat-derived stem cells (OFSCs) possessed multi-lineage differentiation potential to become osteoblasts, chondrocytes, and adipocytes. Upon mix-culture with corneal epithelial cells, OFSCs changed their morphology to round, polygonal epithelial-like cells. Loss of CD105 expression and increased expression of epithelial cell markers, including epithelial-specific antigen and zonal occludin-1, were found upon mix-culture with corneal epithelial cells. Moreover, corneal epithelial differentiation was evidenced by expression of cytokeratin -19 and cytokeratin -3 after mix-culture with corneal epithelial cells, whereas human adipose-derived stem cells from subcutaneous fat were unable to differentiate into corneal epithelial cells under the same induction condition. We further found that direct contact with corneal epithelial cells was essential for OFSCs to commit to corneal epithelial cells. Taken together, orbital fat tissues are a novel source for multi-potent stem cells that possess the potential to differentiate into corneal epithelial lineage. OFSCs are therefore a potential candidate for cell therapy and tissue engineering of corneal epithelium.


Asunto(s)
Órbita/citología , Células Madre/citología , Anciano , Diferenciación Celular/fisiología , Células Cultivadas , Condrogénesis/fisiología , Células Epiteliales/citología , Femenino , Humanos , Inmunohistoquímica , Masculino
5.
J Orthop Res ; 28(1): 131-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19637215

RESUMEN

Change of actin filament organization at the early stage of cell differentiation directs cell fate commitment of mesenchymal stem cells (MSCs). Thymosin beta-4 (Tbeta(4)), a major G-actin sequestering peptide, is known to regulate the cytoskeleton. The study investigated the ways in which Tbeta(4) regulates cell fate determination in MSCs upon differentiation induction. It was found that Tbeta(4) decreased F-actin formation, reduced the F-actin/G-actin ratio, and inhibited osteogenic differentiation; such actin reorganization was not associated with the change of Runt-related transcription factor 2 gene expression during early osteogenic induction. Besides, Tbeta(4) reciprocally facilitated adipogenic differentiation. Tbeta(4) treatment was found to up-regulate gene as well as promote surface expression of adipocyte adhesion molecule during early adipogenic differentiation, which accompanied acceleration of adipocyte phenotypic maturation but was not associated with differential expression of peroxisome proliferator-activated receptor gamma during the first week of adipogenic induction. In summary, Tbeta(4) initiated cell fate determination of MSCs through biophysical effects exerted by cytoskeleton reorganization and altered cell-cell adhesion rather than direct regulation of lineage-determining transcriptional factors. Such findings suggest that Tbeta(4), a ubiquitous peptide, may be involved in osteoporosis when its intracellular concentration is elevated. Further investigation of targeting Tbeta(4) for future osteoporosis treatment is warranted.


Asunto(s)
Diferenciación Celular/genética , Hormonas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Timosina/metabolismo , Actinas/efectos de los fármacos , Actinas/genética , Actinas/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Fenómenos Biofísicos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Hormonas/farmacología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , PPAR gamma/efectos de los fármacos , PPAR gamma/genética , PPAR gamma/metabolismo , Timosina/farmacología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA