Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(24)2023 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-38132138

RESUMO

The therapeutic potential of directly reprogrammed neural stem cells (iNSCs) for neurodegenerative diseases relies on reducing the innate tumorigenicity of pluripotent stem cells. However, the heterogeneity within iNSCs is a major hurdle in quality control prior to clinical applications. Herein, we generated iNSCs from human fibroblasts, by transfecting transcription factors using Sendai virus particles, and characterized the expression of iNSC markers. Using immunostaining and quantitative real time -polymerase chain reaction (RT -qPCR), no differences were observed between colonies of iNSCs and iNSC-derived neurons. Unexpectedly, patch-clamp analysis of iNSC-derived neurons revealed distinctive action potential firing even within the same batch product. We performed single-cell RNA sequencing in fibroblasts, iNSCs, and iNSC-derived neurons to dissect their functional heterogeneity and identify cell fate regulators during direct reprogramming followed by neuronal differentiation. Pseudotime trajectory analysis revealed distinct cell types depending on their gene expression profiles. Differential gene expression analysis showed distinct NEUROG1, PEG3, and STMN2 expression patterns in iNSCs and iNSC-derived neurons. Taken together, we recommend performing a predictable functional assessment with appropriate surrogate markers to ensure the quality control of iNSCs and their differentiated neurons, particularly before cell banking for regenerative cell therapy.


Assuntos
Células-Tronco Neurais , Células-Tronco Pluripotentes , Humanos , Neurônios , Células-Tronco Neurais/metabolismo , Diferenciação Celular/genética , Análise de Sequência de RNA
2.
Int J Stem Cells ; 16(3): 281-292, 2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37105557

RESUMO

Background and Objectives: Human induced pluripotent stem cell (hiPSC)-derived cardiomyocyte (CM) hold great promise as a cellular source of CM for cardiac function restoration in ischemic heart disease. However, the use of animal-derived xenogeneic substances during the biomanufacturing of hiPSC-CM can induce inadvertent immune responses or chronic inflammation, followed by tumorigenicity. In this study, we aimed to reveal the effects of xenogeneic substances on the functional properties and potential immunogenicity of hiPSC-CM during differentiation, demonstrating the quality and safety of hiPSC-based cell therapy. Methods and Results: We successfully generated hiPSC-CM in the presence and absence of xenogeneic substances (xeno-containing (XC) and xeno-free (XF) conditions, respectively), and compared their characteristics, including the contractile functions and glycan profiles. Compared to XC-hiPSC-CM, XF-hiPSC-CM showed early onset of myocyte contractile beating and maturation, with a high expression of cardiac lineage-specific genes (ACTC1, TNNT2, and RYR2) by using MEA and RT-qPCR. We quantified N-glycolylneuraminic acid (Neu5Gc), a xenogeneic sialic acid, in hiPSC-CM using an indirect enzyme-linked immunosorbent assay and liquid chromatography-multiple reaction monitoring- mass spectrometry. Neu5Gc was incorporated into the glycans of hiPSC-CM during xeno-containing differentiation, whereas it was barely detected in XF-hiPSC-CM. Conclusions: To the best of our knowledge, this is the first study to show that the electrophysiological function and glycan profiles of hiPSC-CM can be affected by the presence of xenogeneic substances during their differentiation and maturation. To ensure quality control and safety in hiPSC-based cell therapy, xenogeneic substances should be excluded from the biomanufacturing process.

3.
Tissue Cell ; 47(1): 10-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25441616

RESUMO

Human amniotic membrane-derived stromal cells (hAMSC) are candidates for cell-based therapies. We examined the characteristics of hAMSC including the interaction between hAMSC and breast cancer cells, MCF-7, and MDA-MB-231. Human amniotic membrane-derived stromal cells showed typical MSC properties, including fibroblast-like morphology, surface antigen expression, and mesodermal differentiation. To investigate cell-cell interaction via secreted molecules, we cultured breast cancer cells in hAMSC-conditioned medium (hAMSC-CM) and analyzed their proliferation, migration, and secretome profiles. MCF-7 and MDA-MB-231 cells exposed to hAMSC-CM showed increased proliferation and migration. However, in hAMSC-CM, MCF-7 cells proliferated significantly faster than MDA-MB-231 cells. When cultured in hAMSC-CM, MCF-7 cells migrated faster than MDA-MB-231 cells. Two cell types showed different profiles of secreted factors. MCF-7 cells expressed much amounts of IL-8, GRO, and MCP-1 in hAMSC-CM. Human amniotic membrane-derived stromal cells interact with breast cancer cells through secreted molecules. Factors secreted by hAMSCs promote the proliferation and migration of MCF-7 breast cancer cells. For much safe cell-based therapies using hAMSC, it is necessary to study carefully about interaction between hAMSC and cancer cells.


Assuntos
Âmnio/citologia , Neoplasias da Mama/genética , Proliferação de Células/genética , Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos , Âmnio/metabolismo , Neoplasias da Mama/patologia , Movimento Celular/genética , Quimiocina CCL2/biossíntese , Quimiocina CXCL1/biossíntese , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-8/biossíntese , Células MCF-7 , Células Estromais
4.
Neurol Res ; 36(3): 215-23, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24512015

RESUMO

Even though erythropoietin (EPO) is a neurotropic cytokine that is recognized widely for its role in the development, maintenance, protection, and repair of the nervous system, there are few reports concerning EPO-mediated influences on the glial cells in the central nervous system. In this study, we investigated anti-inflammatory and anti-apoptotic effects of EPO on C6 glioma cells (C6 cells). Erythropoietin did not attenuate inflammatory response, such as nitrite production, iNOS gene expression, and pro-inflammatory cytokines when LPS/TNF-alpha mixture was treated. However, EPO increased C6 cell viability by exerting cell protective effect against staurosporine stimulation. Erythropoietin increased the transient Akt expression at 30 minutes and induced the gradual elevation of ERK1/2 and p38 expression as time progressed. The cell protective effect of EPO was also significantly attenuated with pretreatment of specific PI3K, pERK1/2, or pP38 inhibitor. In summary, these results suggest that EPO may exert its cell protective functions via the direct cell protective activity rather than via its anti-inflammatory effect. Moreover, the PI3K/Akt and mitogen activated protein kinase (MAPK) pathways may be responsible for cell survival against cytotoxicity.


Assuntos
Eritropoetina/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Eritropoetina/metabolismo , Mediadores da Inflamação/metabolismo , Neuroglia/metabolismo , Ratos
5.
Arch Pharm Res ; 36(8): 1013-22, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23615814

RESUMO

Human mesenchymal stem cells (hMSCs) have generated a great deal of interest in clinical application due to their ability to undergo multi-lineage differentiation. Recently, ex vivo genetic modification of hMSCs was attempted to increase their differentiation potential. The present study was conducted to evaluate the biodistribution and in vivo efficacy of genetically modified hMSCs. To accomplish this, Runx2, which is a key transcription factor associated with osteoblast differentiation, was transduced into hMSCs using lentiviral vectors expressing green fluorescent protein (GFP) or luciferase. Here, we developed an experimental fracture in mice femur to investigate the effects of Runx2-transduced hMSCs on bone healing and migration into injury site. We conducted bio-luminescence imaging (BLI) using luciferase-tagged vector and quantitative real-time PCR using GFP probe to investigate the biodistribution of Runx2-transduced hMSCs in the fracture model. The biodistribution of hMSC cells in the fractured femur was observed at 14 days post-transplantation upon both BLI imaging and real-time PCR. Moreover, the fractured mice transplanted with Runx2-transduced hMSCs showed superior bone healing when compared to mock-transduced hMSC and MRC5 fibroblasts which were used as control. These data suggested that transplanted genetically modified hMSCs systemically migrate to the fractured femur, where they contribute to bone formation in vivo.


Assuntos
Modelos Animais de Doenças , Fêmur/lesões , Fraturas Ósseas/genética , Fraturas Ósseas/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Células Cultivadas , Fêmur/metabolismo , Fraturas Ósseas/metabolismo , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Distribuição Tecidual/fisiologia
6.
Toxicol Mech Methods ; 22(2): 118-30, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22082211

RESUMO

We developed and analyzed a new surrogate endpoint of the mouse embryonic stem cell test (EST) for developmental neurotoxicity. To determine the sensitivity, specificity, and transferability of the new endpoint, a pre-validation team from three independent laboratories optimized and standardized the protocol for neuronal differentiation of mouse embryonic stem cells (mESCs) by measuring the neuronal differentiation rates of mESCs under different culture conditions, such as the presence or absence of basic fibroblast growth factor (bFGF) in the growth media and varying lengths of culture. In addition, a component ratio of neuronal cells was measured by using flow cytometry analysis of ß-III tubulin (Tuj1)-positive cells and real-time polymerase chain reaction analysis of microtubule-associated protein 2 (MAP2) mRNA. Our results showed that the best growth was achieved by culturing mESCs for 12 d in N2B27 medium without bFGF or ascorbic acid. Lead (II) acetate and aroclor 1254 were used to test the usefulness of the new endpoint. When we used the known ID(50) values for lead (II) acetate in the EST model, it was classified as non-embryotoxic; however, when we used the new ID(50) values that we determined in this study, it was classified as weakly embryotoxic. Aroclor 1254 and penicillin G were also classified as weakly embryotoxic and non-embryotoxic compounds, respectively, when cardiac and neuronal differentiation ID(50) values were used. Therefore, our new surrogate endpoint for developmental neurotoxicity is not only sensitive and specific but also transferable among laboratories.


Assuntos
Células-Tronco Embrionárias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Chumbo/toxicidade , Camundongos , Células NIH 3T3
7.
Int J Toxicol ; 30(5): 498-509, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22013134

RESUMO

We developed a new end point of the mouse stem cell test (EST) for developmental neurotoxicity. We tested 2 developmental neurotoxicants, namely, lead (II) acetate and Aroclor 1254, using this EST. Our results showed that lead (II) acetate is nonembryotoxic, and Aroclor 1254 is weakly embryotoxic. To identify a new end point for developmental neurotoxicity, we used the default method of neuronal differentiation for D3 mouse embryonic stem cells with basic fibroblast growth factor (bFGF) and ascorbic acid. Flow cytometry and real-time polymerase chain reaction were used to quantify the inhibition of neuronal differentiation. Our results showed that both lead (II) acetate and Aroclor 1254 reduced the percentage of microtubule-associated protein 2 (MAP-2)-positive cells and the messenger RNA (mRNA) expression level of MAP-2 in a dose-dependent manner. These results suggested that these methods can be used to develop an additional end point of the EST for developmental neurotoxicity using default differentiation of mouse embryonic stem cells.


Assuntos
/toxicidade , Células-Tronco Embrionárias/efeitos dos fármacos , Determinação de Ponto Final , Compostos Organometálicos/toxicidade , Teratogênicos/toxicidade , Células 3T3 , Animais , Ácido Ascórbico/metabolismo , Diferenciação Celular , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibroblastos/citologia , Citometria de Fluxo , Expressão Gênica , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , RNA Mensageiro , Reação em Cadeia da Polimerase em Tempo Real
8.
Stem Cells Dev ; 20(1): 149-57, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20486780

RESUMO

Embryonic stem cell (ESC) research gave rise to the possibility that stem cell therapy could be used in the treatment of incurable diseases such as neurodegenerative disorders. However, problems related to the tumorigenicity of undifferentiated ESCs must be resolved before such cells can be used in the application of cell replacement therapies. In the present study, we attempted to determine biomarkers that predicted tumor formation of undifferentiated ESCs in vivo. We differentiated mouse ESCs (R1 cell line) into neural lineage using a 5-step method, and evaluated the expression of oncogenes (p53, Bax, c-myc, Bcl2, K-ras), telomerase-related genes (TERT, TRF), and telomerase activity and telomere length during differentiation of ESCs. The expression of oncogenes did not show a significant change during differentiation steps, but the expression of telomerase reverse transcriptase (TERT) and telomerase activity correlated with mouse ESCs differentiation. To investigate the possibility of mouse TERT (mTERT) as a biomarker of tumorigenicity of undifferentiated ESCs, we established mTERT knockdown ESCs using the shRNA lentivirus vector and evaluated its tumorigenicity in vivo using nude mice. Tumor volumes significantly decreased, and appearances of tumor formation in mice were delayed in the TERT-knockdown ESC treated group compared with the undifferentiated ESC treated group. Altogether, these results suggested that mTERT might be potentially beneficial as a biomarker, rather than oncogenes of somatic cells, for the assessment of ESCs tumorigenicity.


Assuntos
Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/patologia , Neoplasias/patologia , Telomerase/metabolismo , Animais , Diferenciação Celular/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/citologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Telômero/metabolismo
9.
Toxicol Res ; 27(4): 247-51, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24278579

RESUMO

To clarify whether inhibitory effect of estrogen on liver tumor is associated with cell proliferation, we investigated its role in diethylnitrosamine (DEN)-induced rat preneoplastic lesions, with time sequenced manners. F344 male rats (n = 90) were divided into three groups at 5 weeks of age. The mini-osmotic pumps providing a continuous infusion of DEN was implanted into the abdominal cavity of each animal in group 1, 2 and 3 at 6 weeks of age. To see the effect of estrogen, pellet containing 1 or 10 µg of estradiol- 3-benzoate (EB) was implanted subcutaneously in the animals of groups 2 or 3, respectively, one week prior to DEN treatment. Ten animals of each group were euthanized at 10, 14 and 18 weeks after DEN treatment. Liver tissues at each time point were fixed in 10% phosphate-buffered formalin and were processed and embedded in paraffin and 5 µm sections mounted on a silanized slide. Glutathione S-transferase placental form (GST-P) positive foci and 5-bromo-2-deoxyuridine (BrdU) labeling cells were detected at each time point. Area of GST-P positive foci in DEN+EB 1 or 10 µg group was significantly decreased compared to DEN alone at 14 weeks (p < 0.01 or p < 0.05, respectively) an at 18 weeks (p < 0.05 or p < 0.01, respectively). BrdU index in DEN+EB 1 or 10 µg groups was significantly decreased compared to DEN alone at 14 weeks and at 18 weeks (p < 0.01). Taken together, we conclude that EB treatment decrease the DEN-induced liver preneoplastic lesions and this may be associated with decrease of cellular proliferation.

10.
Arch Pharm Res ; 32(1): 117-26, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19183884

RESUMO

Human mesenchymal stem cells (hMSCs) have generated a great deal of interest in clinical applications. The reason is that they may have the plasticity needed to differentiate into multiple lineages and the ability to expand ex vivo. For the therapeutic applications of hMSCs to be of practical use, it is crucial to assess the efficacy and safety of hMSCs in long-term ex vivo expansion. In this study, we cultured hMSCs by population doubling (PD) 60, and investigated their growth, osteogenic and adipogenic differential abilities, change of surface markers, telomerase activity, telomere length, and gene expression related to tumorigenesis. An in vivo tumorigenesis assay was also carried out. In long-term expanded hMSCs, the cells became aged above PD 30 and their adipogenic and osteogenic differentiation potential decreased. Telomerase activity unchanged whereas telomere length decreased and karyotypes were not changed. Gene expressions related to tumorigenesis decreased in proportion as the PD of hMSCs increased. In vivo transplantation of long-term cultured hMSCs to nude mice did not result in tumor formation. These findings suggest that diverse tests for cellular therapy should be considered during the ex vivo culture of hMSCs, particularly when a prolonged and extended propagation period is required.


Assuntos
Diferenciação Celular , Proliferação de Células , Células-Tronco Mesenquimais/fisiologia , Adipogenia , Animais , Forma Celular , Transformação Celular Neoplásica/genética , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Osteócitos/fisiologia , Fenótipo , Telomerase/metabolismo , Telômero/metabolismo , Fatores de Tempo
11.
Biochem Biophys Res Commun ; 366(3): 624-30, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18047835

RESUMO

The molecular mechanisms that couple growth arrest and cell differentiation were examined during adipogenesis. Here, to understand the cyclin-dependent kinase inhibitor (CKI) genes involved in the progression of adipogenic differentiation, we examined changes in the protein and mRNA expression levels of CKI genes in vitro. During the onset of growth arrest associated with adipogenic differentiation, two independent families of CKI genes, p27Kip1 and p18INK4c, were significantly increased. The expressions of p27Kip1 and p18INK4c, regulated at the level of protein and mRNA accumulation, were directly coupled to adipogenic differentiation. This finding was supported by the inhibition of adipogenic differentiation caused by short interfering RNA (siRNA). In this study, we investigated the regulatory effects of transforming growth factor beta-1 (TGFbeta-1) on CKI genes involved in adipogenic differentiation of bone marrow-derived human mesenchymal stem cells (hMSCs). Only the up-regulation of p18INK4c during adipogenic differentiation, and not that of the p27Kip1 gene was prevented by treatment with TGFbeta-1, one of the factors that inhibit adipogenesis in vitro. This finding indicates a close correlation between adipogenic differentiation and p18INK4c induction in hMSCs. Thus, these data demonstrate a role for the differentiation-dependent cascade expression of cyclin-dependent kinase inhibitors in regulating adipogenic differentiation, thereby providing a molecular mechanism that couples growth arrest and differentiation.


Assuntos
Adipócitos/citologia , Adipócitos/fisiologia , Adipogenia/fisiologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Quinases Ciclina-Dependentes/metabolismo , Humanos
12.
J Vet Sci ; 3(4): 303-14, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12819380

RESUMO

The regional distribution and relative frequency of some endocrine cells in the pancreas of the carp, Cyprinus carpio Linnaeus, belonging to the family Cyprinidae in the order Cypriniformes, were observed using specific mammalian antisera against insulin, glucagon, somatostatin and human pancreatic polypeptide (hPP) by peroxidase antiperoxidase (PAP) method. The pancreas was divided into four regions (principal and secondary islets, exocrine and pancreatic duct regions). In addition, the pancreatic islet regions were further subdivided into three regions (central, mantle and peripheral regions) and the pancreatic duct regions were subdivided into two regions (epithelial and subepithelial regions). Spherical to spindle or occasionally round to oval shaped immunoreactive (IR) cells were demonstrated in the pancreatic islets, exocrine and pancreatic duct. In the principal islet regions, some cells were also detected in the other regions, most of insulin- and somatostatin-IR cells were located in the central regions, and glucagon- and hPP-IR cells were situated in the peripheral regions. In this regions, insulin-IR cells were most predominant cell types and then, glucagon, somatostatin and hPP in that order. In the secondary islet regions, the regional distribution and relative frequency of these four types of endocrine cells were quite similar to those of the principal islets except for cell clusters consisted of hPP-IR cells that were situated in the peripheral to mantle regions. In the pancreatic duct regions, all four major pancreatic endocrine cells were demonstrated in the inter-epithelial cells and/or basal regions of the epithelial linning. In addition, cell clusters composed of numerous insulin-, moderate glucagon- and somatostatin-IR cells of low frequency were also observed in the subepithelial regions of the pancreatic duct. In the exocrine regions, insulin-, glucagon-, somatostatin- and hPP-IR cells were located in the inter-acinus regions with rare, a few, moderate and moderate frequencies, respectively. In conclusion, the regional distribution and relative frequency of four major pancreatic endocrine cells, insulin-, glucagon-, somatostatin- and hPP-IR cells, in the pancreas of the carp showed general patterns which were observed in other stomachless teleost. However, some species- dependent different distributional patterns and/or relative frequencies were also demonstrated.


Assuntos
Carpas/metabolismo , Pâncreas/metabolismo , Animais , Feminino , Glucagon/metabolismo , Imuno-Histoquímica/veterinária , Insulina/metabolismo , Masculino , Pâncreas/citologia , Polipeptídeo Pancreático/metabolismo , Somatostatina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...