Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Stem Cells ; 38(4): 516-529, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31778275

RESUMEN

Pluripotent stem cells (PSCs) can serve as an unlimited cell source for transplantation therapies for treating various devastating diseases, such as cardiovascular diseases, diabetes, and Parkinson's disease. However, PSC transplantation has some associated risks, including teratoma formation from the remaining undifferentiated PSCs. Thus, for successful clinical application, it is essential to ablate the proliferative PSCs before or after transplantation. In this study, neural stem cell-derived conditioned medium (NSC-CM) inhibited the proliferation of PSCs and PSC-derived neural precursor (NP) cells without influencing the potential of PSC-NP cells to differentiate into neurons in vitro and prevented teratoma growth in vivo. Moreover, we found that the NSC-CM remarkably decreased the expression levels of Oct4 and cyclin D1 that Oct4 directly binds to and increased the cleaved-caspase 3-positive cell death through the DNA damage response in PSCs and PSC-NPs. Interestingly, we found that NSCs distinctly secreted the tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2 proteins. These proteins suppressed not only the proliferation of PSCs in cell culture but also teratoma growth in mice transplanted with PSCs through inhibition of matrix metalloproteinase (MMP)-2 and MMP-9 activity. Taken together, these results suggest that the TIMP proteins may improve the efficacy and safety of the PSC-based transplantation therapy.


Asunto(s)
Células Madre Pluripotentes/metabolismo , Teratoma/terapia , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Animales , Humanos , Masculino , Ratones , Ratones Desnudos , Teratoma/patología
2.
Biochem Biophys Res Commun ; 518(2): 388-395, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31421823

RESUMEN

UV-induced skin damage is involved in ROS overproduction and the overexpression of matrix metalloproteinases (MMPs), which are inhibited by TIMPs (tissue inhibitor of neural stem cells (NSCs)). These proteins may be associated with skin regeneration through the activation of TIMP proteins, but there have been no reports of treatment of skin photodamage using NSCs and their secreted proteins TIMP-1 and TIMP-2. Here we investigated the photoprotective role of NSCs and their TIMP proteins for the inhibition of UVB-irradiation damage in fibroblasts in SKH-1 mice. SKH-1 hairless mice were divided into three groups (n = 4 per group): normal, treatment, and control groups. The latter two groups were dorsally exposed to UVB irradiation for 12 weeks. After UVB irradiation, treatments with NSC-CM and its secreted factors TIMP-1 and TIMP-2, markedly ameliorated the photodamage triggered by the increase in MMP expression and activity through ROS production, and the subsequent activation of the NF-κB pathway in UVB-irradiated fibroblasts and the treatment mouse group. In addition, the topical application of NSC-CM to mice in the treatment group after irradiation clearly inhibited the expression of γ-H2AX, a DNA damage marker, through the activation of the DNA repair enzyme Rad50. These results demonstrate that NSC-CM or TIMPs proteins can ameliorate skin photodamage induced by UVB-irradiation in in vitro and in vivo systems.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Células-Madre Neurales/metabolismo , Envejecimiento de la Piel/efectos de los fármacos , Inhibidores Tisulares de Metaloproteinasas/farmacología , Rayos Ultravioleta/efectos adversos , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Línea Celular , Medios de Cultivo Condicionados/metabolismo , Femenino , Humanos , Ratones , Ratones Pelados , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Piel/efectos de los fármacos , Piel/metabolismo , Piel/efectos de la radiación , Envejecimiento de la Piel/efectos de la radiación , Inhibidores Tisulares de Metaloproteinasas/metabolismo
3.
Methods ; 133: 104-112, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28867501

RESUMEN

Therapeutic approaches based on stem cells have received considerable attention as potential treatments for Huntington's disease (HD), which is a fatal, inherited neurodegenerative disorder, caused by progressive loss of GABAergic medium spiny neurons (MSNs) in the striatum of the forebrain. Transplantation of stem cells or their derivatives in animal models of HD, efficiently improved functions by replacing the damaged or lost neurons. In particular, neural stem cells (NSCs) for HD treatments have been developed from various sources, such as the brain itself, the pluripotent stem cells (PSCs), and the somatic cells of the HD patients. However, the brain-derived NSCs are difficult to obtain, and the PSCs have to be differentiated into a population of the desired neuronal cells that may cause a risk of tumor formation after transplantation. In contrast, induced NSCs, derived from somatic cells as a new stem cell source for transplantation, are less likely to form tumors. Given that the stem cell transplantation strategy for treatment of HD, as a genetic disease, is to replace the dysfunctional or lost neurons, the correction of mutant genes containing the expanded CAG repeats is essential. In this review, we will describe the methods for obtaining the optimal NSCs for transplantation-based HD treatment and the differentiation conditions for the functional GABAergic MSNs as therapeutic cells. Also, we will discuss the valuable gene correction of the disease stem cells by the CRISPR/Cas9 system for HD treatment.


Asunto(s)
Enfermedad de Huntington/terapia , Células Madre Pluripotentes Inducidas/trasplante , Células-Madre Neurales/trasplante , Trasplante de Células Madre/tendencias , Animales , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Humanos , Neuronas/patología , Neuronas/trasplante
4.
Biochem Biophys Res Commun ; 476(1): 42-8, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27207831

RESUMEN

The generation of induced neural stem cells (iNSCs) from somatic cells using defined factors provides new avenues for basic research and cell therapies for various neurological diseases, such as Parkinson's disease, Huntington's disease, and spinal cord injuries. However, the transcription factors used for direct reprogramming have the potential to cause unexpected genetic modifications, which limits their potential application in cell therapies. Here, we show that a combination of four chemical compounds resulted in cells directly acquiring a NSC identity; we termed these cells chemically-induced NSCs (ciNSCs). ciNSCs expressed NSC markers (Pax6, PLZF, Nestin, Sox2, and Sox1) and resembled NSCs in terms of their morphology, self-renewal, gene expression profile, and electrophysiological function when differentiated into the neuronal lineage. Moreover, ciNSCs could differentiate into several types of mature neurons (dopaminergic, GABAergic, and cholinergic) as well as astrocytes and oligodendrocytes in vitro. Taken together, our results suggest that stably expandable and functional ciNSCs can be directly reprogrammed from mouse fibroblasts using a combination of small molecules without any genetic manipulation, and will provide a new source of cells for cellular replacement therapy of neurodegenerative diseases.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Reprogramación Celular/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Animales , Diferenciación Celular , Línea Celular , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo
5.
ScientificWorldJournal ; 2014: 957548, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24991651

RESUMEN

Somatic cells were directly converted to functional neurons through the use of a combination of transcription factors, including Ascl1, Brn2, and Myt1l. However, a major limitation is the lack of a reliable source of cell-replacement therapy for neurological diseases. Here, we show that a combination of the transcription factors Ascl1 and Nurr1 (AN) and neurotrophic factors including SHH and FGF8b directly reprogrammed embryonic mouse fibroblasts to induced neuronal (iN) cells: pan-neuronal cells and dopaminergic (DA) neurons under our systematic cell culture conditions. Reprogrammed cells showed the morphological properties of neuronal cells. Additionally, cells were analyzed using various markers, including Tuj1 and Map2 for neuronal cells and Lmx1a, Th, Aadc and Vmat2 for DA neurons in our immunostaining and reverse transcription (RT)-PCR experiments. We found that a combination of transcription factors and neurotrophic factors could directly reprogram fibroblasts to neuronal cells including DA neurons. Various types of reprogrammed cells are promising cell sources for cell-based therapy of neurological disorders like Parkinson's disease and spinal cord injury.


Asunto(s)
Reprogramación Celular/fisiología , Neuronas Dopaminérgicas/fisiología , Fibroblastos/fisiología , Células-Madre Neurales/fisiología , Animales , Humanos , Ratones , Ratones Endogámicos BALB C , Neuronas/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología
6.
J Cell Biochem ; 114(4): 920-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23097321

RESUMEN

Establishment of mouse spermatogonial stem cell (SSC) culture systems offers a useful stem cell model for studies of proliferation and self-renewal of mammalian germline stem cells. In addition, spontaneous development of pluripotent stem cells from cultured SSCs emphasizes their possible applications in regenerative medicine as a substitute for embryonic stem cells (ESCs). These pluripotent stem cells termed multipotent germline stem cells (mGSCs) or germline-derived pluripotent stem cells (gPSCs) exhibit almost identical properties in terms of morphology and gene expression patterns to mouse ESCs (mESCs). In this study, to help understand mechanisms underlying reprogramming of SSCs into pluripotent stem cells, we established a culture system of SSCs derived from mice harboring green fluorescence protein (GFP) transgene whose expression is modulated by Oct4 regulatory sequences. Our results indicated that GFP intensity faithfully reflected cellular states upon reprogramming of SSCs or treatment with a selective extracellular signal-regulated kinase (ERK) inhibitor PD0325901. Moreover, in contrast to mESCs, regulation of Nanog expression did not appear to couple to the Oct4 level in SSCs. Further analysis of Oct4-GFP SSCs demonstrated that a posttranscriptional control of pluripotency marker genes such as Oct4 and Sox2 might play an important role as an inhibitory mechanism preventing the acquisition of pluripotency.


Asunto(s)
Reprogramación Celular , Células Madre Embrionarias/citología , Proteínas Fluorescentes Verdes/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Animales , Benzamidas/farmacología , Diferenciación Celular , Forma de la Célula , Células Cultivadas , Difenilamina/análogos & derivados , Difenilamina/farmacología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Fibroblastos/efectos de la radiación , Rayos gamma , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas de Homeodominio/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Ratones Transgénicos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Procesamiento Proteico-Postraduccional , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Testículo/citología , Testículo/metabolismo , Transgenes
8.
NPJ Regen Med ; 7(1): 4, 2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35027563

RESUMEN

The generation of human oligodendrocyte progenitor cells (OPCs) may be therapeutically valuable for human demyelinating diseases such as multiple sclerosis. Here, we report the direct reprogramming of human somatic cells into expandable induced OPCs (iOPCs) using a combination of OCT4 and a small molecule cocktail. This method enables generation of A2B5+ (an early marker for OPCs) iOPCs within 2 weeks retaining the ability to differentiate into MBP-positive mature oligodendrocytes. RNA-seq analysis revealed that the transcriptome of O4+ iOPCs was similar to that of O4+ OPCs and ChIP-seq analysis revealed that putative OCT4-binding regions were detected in the regulatory elements of CNS development-related genes. Notably, engrafted iOPCs remyelinated the brains of adult shiverer mice and experimental autoimmune encephalomyelitis mice with MOG-induced 14 weeks after transplantation. In conclusion, our study may contribute to the development of therapeutic approaches for neurological disorders, as well as facilitate the understanding of the molecular mechanisms underlying glial development.

9.
Artif Cells Nanomed Biotechnol ; 48(1): 1303-1313, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33150801

RESUMEN

Cancer stem cells (CSCs) have been implicated in the growth and progression of several types of human cancer. The technology to derive and establish CSCs in vitro could be a critical tool for understanding cancer and developing new therapeutic targets. In this study, we derived expandable CD15+ induced CSCs (iCSCs) from immortalised 293FT human epithelial cells by co-culture with human bone marrow-derived mesenchymal stem cells (BM-MSCs) as feeder cells in vitro. The iCSCs converted through an epithelial-mesenchymal transition program acquired mesenchymal traits, the expression of stem cell markers, and epigenetic changes. Moreover, the iCSCs not only efficiently formed tumorspheres in vitro but also initiated tumours in immunocompromised mice injected with only 10 of the iCSCs. Furthermore, we showed that the expression of the chemokine CXCL12 and its receptor CXCR4 by the iCSCs resulted in the activation of the Fut4 gene through CXCR4/ERK/ELK-1-signalling pathways and the maintenance of the iCSCs in the undifferentiated state through CXCR4/AKT/STAT3-signalling. These findings suggest that immortalised 293FT cells may acquire potential oncogenicity through molecular and cellular alteration processes in microenvironments using BM-MSCs, and could represent a valuable in vitro model as a cancer stem cell surrogate for studying the pathophysiological properties of CSCs.


Asunto(s)
Quimiocina CXCL12/metabolismo , Células Madre Neoplásicas/patología , Receptores CXCR4/metabolismo , Transducción de Señal , Animales , Carcinogénesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Ratones , Proteína Elk-1 con Dominio ets/metabolismo
10.
Artif Cells Nanomed Biotechnol ; 46(sup1): 1114-1122, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29506416

RESUMEN

Direct conversion is a powerful approach to safely generate mature neural lineages with potential for treatment of neurological disorders. Astrocytes play a crucial role in neuronal homeostasis and their dysfunctions contribute to several neurodegenerative diseases. Using a single-cell approach for precision, we describe here a robust method using optimized DNA amounts for the direct conversion of mouse fibroblasts to astrocytes. Controlled amount of the reprogramming factors Oct4, Sox2, Klf4 and cMyc was directly delivered into a single fibroblast cell. Consequently, 2500 DNA molecules, no more or less, were found to be the optimal amount that dramatically increased the expression levels of the astrocyte-specific markers GFAP and S100b and the demethylation gene TET1, the expression of which was sustained to maintain astrocyte functionality. The converted astrocytes showed glutamate uptake ability and electrophysiological activity. Furthermore, we demonstrated a potential mechanism whereby fibroblast was directly converted into astrocyte at a single-cell level; this was achieved by activating BMP2 pathway through direct binding of Sox2 protein to BMP2 gene. This study suggests that nanotechnology for directly injecting plasmid DNAs into cell nuclei may help understand such a conversion at single-cell level.


Asunto(s)
Astrocitos/citología , ADN/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Fibroblastos/citología , Fibroblastos/metabolismo , Nanotecnología/métodos , Plásmidos/genética , Animales , Secuencia de Bases , Linaje de la Célula , ADN/genética , ADN/metabolismo , Inyecciones , Factor 4 Similar a Kruppel , Ratones
11.
Expert Opin Biol Ther ; 17(11): 1333-1343, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28792249

RESUMEN

INTRODUCTION: Huntington's disease (HD) is an inherited neurodegenerative disease characterized by chorea, dementia, and depression caused by progressive nerve cell degeneration, which is triggered by expanded CAG repeats in the huntingtin (Htt) gene. Currently, there is no cure for this disease, nor is there an effective medicine available to delay or improve the physical, mental, and behavioral severities caused by it. Areas covered: In this review, the authors describe the use of induced neural stem cells (iNSCs) by direct conversion technology, which offers great advantages as a therapeutic cell type to treat HD. Expert opinion: Cell conversion of somatic cells into a desired stem cell type is one of the most promising treatments for HD because it could be facilitated for the generation of patient-specific neural stem cells. The induced pluripotent stem cells (iPSCs) have a powerful potential for differentiation into neurons, but they may cause teratoma formation due to an undifferentiated pluripotent stem cell after transplantation Therefore, direct conversion of somatic cells into iNSCs is a promising alternative technology in regenerative medicine and the iNSCs may be provided as a therapeutic cell source for Huntington's disease.


Asunto(s)
Enfermedad de Huntington/terapia , Células Madre Pluripotentes Inducidas/trasplante , Diferenciación Celular , Humanos , Enfermedad de Huntington/patología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Teratoma/etiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Cell Transplant ; 25(3): 593-607, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26407027

RESUMEN

Neuropathic pain following spinal cord injury (SCI) is a devastating disease characterized by spontaneous pain such as hyperalgesia and allodynia. In this study, we investigated the therapeutic potential of ESC-derived spinal GABAergic neurons to treat neuropathic pain in a SCI rat model. Mouse embryonic stem cell-derived neural precursor cells (mESC-NPCs) were cultured in media supplemented with sonic hedgehog (SHH) and retinoic acid (RA) and efficiently differentiated into GABAergic neurons. Interestingly, low doses of SHH and RA induced MGE-like progenitors, which expressed low levels of DARPP32 and Nkx2.1 and high levels of Irx3 and Pax6. These cells subsequently generated the majority of the DARPP32(-) GABAergic neurons after in vitro differentiation. The spinal mESC-NPCs were intrathecally transplanted into the lesion area of the spinal cord around T10-T11 at 21 days after SCI. The engrafted spinal GABAergic neurons remarkably increased both the paw withdrawal threshold (PWT) below the level of the lesion and the vocalization threshold (VT) to the level of the lesion (T12, T11, and T10 vertebrae), which indicates attenuation of chronic neuropathic pain by the spinal GABAergic neurons. The transplanted cells were positive for GABA antibody staining in the injured region, and cells migrated to the injured spinal site and survived for more than 7 weeks in L4-L5. The mESC-NPC-derived spinal GABAergic neurons dramatically attenuated the chronic neuropathic pain following SCI, suggesting that the spinal GABAergic mESC-NPCs cultured with low doses of SHH and RA could be alternative cell sources for treatment of SCI neuropathic pain by stem cell-based therapies.


Asunto(s)
Células Madre Embrionarias/citología , Neuronas GABAérgicas/trasplante , Células-Madre Neurales/trasplante , Neuralgia/etiología , Neuralgia/terapia , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/terapia , Animales , Línea Celular , Neuronas GABAérgicas/citología , Masculino , Ratones , Células-Madre Neurales/citología , Neuralgia/patología , Neurogénesis , Umbral del Dolor , Ratas Sprague-Dawley , Médula Espinal/patología , Traumatismos de la Médula Espinal/patología
13.
Cell Transplant ; 25(8): 1439-51, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27110030

RESUMEN

Several types of hair loss result from the inability of hair follicles to initiate the anagen phase of the hair regeneration cycle. Modulating signaling pathways in the hair follicle niche can stimulate entry into the anagen phase. Despite much effort, stem cell-based or pharmacological therapies to activate the hair follicle niche have not been successful. Here, we set out to test the effect of neural stem cell (NSC) extract on the hair follicle niche for hair regrowth. NSC extracts were applied to the immortalized cell lines HaCaT keratinocytes and dermal papilla cells (DPCs) and the shaven dorsal skin of mice. Treatment with NSC extract dramatically improved the growth of HaCaT keratinocytes and DPCs. In addition, NSC extract enhanced the hair growth of the shaven dorsal skin of mice. In order to determine the molecular signaling pathways regulated by NSCs, we evaluated the expression levels of multiple growth and signaling factors, such as insulin-like growth factor-1 (IGF-1), hepatocyte growth factor (HGF), keratinocyte growth factor (KGF), vascular endothelial growth factor (VEGF), transforming growth factor-ß (TGF-ß), and bone morphogenetic protein (BMP) family members. We found that treatment with an NSC extract enhanced hair growth by activating hair follicle niches via coregulation of TGF-ß and BMP signaling pathways in the telogen phase. We also observed activation and differentiation of intrafollicular hair follicle stem cells, matrix cells, and extrafollicular DPCs in vivo and in vitro. We tested whether activation of growth factor pathways is a major effect of NSC treatment on hair growth by applying the growth factors to mouse skin. Combined growth factors, including TGF-ß, significantly increased the hair shaft length and growth rate. DNA damage and cell death were not observed in skin cells of mice treated with the NSC extract for a prolonged period. Overall, our data demonstrate that NSC extract provides an effective approach for promoting hair growth by directly regulating hair follicle niches through TGF-ß and BMP signaling pathways as well as induction of core growth factors.


Asunto(s)
Folículo Piloso/citología , Folículo Piloso/metabolismo , Cabello/citología , Cabello/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células Cultivadas , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Biomaterials ; 39: 47-58, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25477171

RESUMEN

The generation of induced pluripotent stem cells (iPSCs) often causes genetic and epigenetic defects, which may limit their clinical applications. Here, we show that reprogramming in the presence of small molecules preserved the genomic stability of iPSCs by inhibiting DNA double-strand breaks (DSBs) and activating Zscan4 gene. Surprisingly, the small molecules protected normal karyotype by facilitating repair of the DSBs that occurred during the early reprogramming process and long-term culture of iPSCs. The stemness and cell growth of iPSCs(+) were normally sustained with high expression of pluripotency genes compared that of iPSCs(-). Moreover, small molecules maintained the differentiation potential of iPSCs(+) for the three germ layers, whereas it was lost in iPSCs(-). Our results demonstrate that the defined small molecules are potent factors for generation of high quality iPSCs with preservation of genomic integrity by facilitating the reprogramming process.


Asunto(s)
Inestabilidad Genómica/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Animales , Ácido Ascórbico/farmacología , Benzamidas/farmacología , Western Blotting , Células Cultivadas , Roturas del ADN de Doble Cadena/efectos de los fármacos , Dioxoles/farmacología , Difenilamina/análogos & derivados , Difenilamina/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Citometría de Flujo , Inestabilidad Genómica/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Pirimidinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiazoles/farmacología
15.
Biotechnol J ; 9(7): 882-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24827816

RESUMEN

Huntington's disease (HD) is a fatal inherited neurodegenerative disorder characterized by progressive loss of neurons in the striatum, a sub-cortical region of the forebrain. The sub-cortical region of the forebrain is associated with the control of movement and behavior, thus HD initially presents with coordination difficulty and cognitive decline. Recent reprogramming technologies, including induced pluripotent stem cells (iPSCs) and induced neural stem cells (iNSCs), have created opportunities to understand the pathological cascades that underlie HD and to develop new treatments for this currently incurable neurological disease. The ultimate objectives of stem cell-based therapies for HD are to replace lost neurons and to prevent neuronal dysfunction and death. In this review, we examine the current understanding of the molecular and pathological mechanisms involved in HD. We discuss disease modeling with HD-iPSCs derived from the somatic cells of patients, which could provide an invaluable platform for understanding HD pathogenesis. We speculate about the benefits and drawbacks of using iNSCs as an alternative stem cell source for HD treatment. Finally, we discuss cell culture and engineering systems that promote the directed differentiation of pluripotent stem cell-derived NSCs into a striatal DARPP32(+) GABAergic MSN phenotype for HD. In conclusion, this review summarizes the potentials of cell reprogramming and engineering technologies relevant to the development of cell-based therapies for HD.


Asunto(s)
Ingeniería Celular/métodos , Enfermedad de Huntington/terapia , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo
16.
J Dermatol Sci ; 72(3): 274-83, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24016750

RESUMEN

BACKGROUND: Melanin for skin pigmentation is synthesized from tyrosine via an enzymatic cascade that is controlled by tyrosinase (TYR), tyrosinase-related protein 1 (TRP1), and dopachrome tautomerase/tyrosinase related protein 2 (Dct/TRP2), which are the targets of microphthalmia-associated transcription factor (MITF). MITF is a master regulator of pigmentation and a target of ß-catenin in Wnt/ß-catenin signaling during melanocyte differentiation. Stem cells have been used in skin pigmentation studies, but the mechanisms were not determined for the conditioned medium (CM)-mediated effects. OBJECTIVES: In this study, the inhibition and mechanisms of melanin synthesis were elucidated in B16 melanoma cells and UV-B irradiated C57/BL-6 mice that were treated with human neural stem cell-conditioned medium (NSC-CM). METHODS: B16-F10 melanoma cells (1.5×10(4)cells/well) and the shaved dorsal skin of mice were pretreated with various amount (5, 10, 20, 50, and 100%) of NSC-CM. Melanin contents and TYR activity were measured by a Spectramax spectrophotometer. The expression of TYR, TRP1, Dct/TRP2, MITF, ß-catenin and Wnt inhibitors were evaluated by RT-PCR and western blot. The dorsal skin samples were analyzed by immunofluorescence with various antibodies and compared with that control of tissues. RESULTS: Marked decreases were evident in melanin content and TYR, TRP1, DCT/TRP2, MITF, and ß-catenin expression in B16 cells and C57/BL-6 mice. NSC-CM negatively regulated Wnt/ß-catenin signaling by decreasing the expression of ß-catenin protein, which resulted from robust expression of Wnt inhibitors Dickkopf-1 (DKK1) and secreted frizzled-related protein 2 (sFRP2). CONCLUSIONS: These results demonstrate that NSC-CM suppresses melanin production in vitro and in vivo, suggesting that factors in NSC-CM may play an important role in deregulation of epidermal melanogenesis.


Asunto(s)
Melaninas/biosíntesis , Melanoma Experimental/metabolismo , Células-Madre Neurales/fisiología , Proteínas Wnt/metabolismo , Animales , Cateninas/metabolismo , Línea Celular Tumoral , Medios de Cultivo Condicionados , Regulación de la Expresión Génica , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Monofenol Monooxigenasa/metabolismo , Pigmentación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA