Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Proc Natl Acad Sci U S A ; 108(31): 12793-8, 2011 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-21768375

RESUMO

Recent evidence indicates that p53 suppression increased the efficiency of induced pluripotent stem cell (iPSC) generation. This occurred even with the enforced expression of as few as two canonical transcription factors, Oct4 and Sox2. In this study, primary human keratinocytes were successfully induced into a stage of plasticity by transient inactivation of p53, without enforced expression of any of the transcription factors previously used in iPSC generation. These cells were later redifferentiated into neural lineages. The gene suppression plastic cells were morphologically indistinguishable from human ES cells. Gene suppression plastic cells were alkaline phosphatase-positive, had normal karyotypes, and expressed p53. Together with the accumulating evidence of similarities and overlapping mechanisms between iPSC generation and cancer formation, this finding sheds light on the emerging picture of p53 sitting at the crossroads between two intricate cellular potentials: stem cell vs. cancer cell generation. This finding further supports the crucial role played by p53 in cellular reprogramming and suggests an alternative method to switch the lineage identity of human cells. This reported method offers the potential for directed lineage switching with the goal of generating autologous cell populations for novel clinical applications for neurodegenerative diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Queratinócitos/metabolismo , Interferência de RNA , Proteína Supressora de Tumor p53/genética , Animais , Western Blotting , Diferenciação Celular/genética , Linhagem da Célula/genética , Transplante de Células/métodos , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Cariotipagem , Queratinócitos/citologia , Camundongos , Camundongos SCID , Camundongos Transgênicos , Microscopia de Fluorescência , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Teratoma/genética , Teratoma/metabolismo , Teratoma/patologia , Transplante Heterólogo , Proteína Supressora de Tumor p53/metabolismo
2.
Transplantation ; 75(5): 679-85, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12640309

RESUMO

BACKGROUND: Mesenchymal stem cells (MSC) are pluripotent progenitors for a variety of cell types, including fibroblasts and muscle cells. Their involvement in the tissue repair of allografts during the development of chronic rejection has been hypothesized, but not yet substantiated, by experimental evidence. METHODS: Rat MSC were isolated from circulation using an aortic pouch allograft as a trapping device. The plasticity of these cells was examined in differentiation cultures. One of the resulting MSC lines was immortalized and transduced to express a marker gene. The -labeled cells were then transferred to F344 rats bearing Lewis (LEW) cardiac allografts to measure their localization and contribution to graft tissue repair. RESULTS: The MSC isolated from circulation exhibited multipotential for differentiation in culture, developing into various lineages including osteoblasts, lipocytes, chondrocytes, myotubes, and fibroblasts. Intravenous engraftment of the -labeled cells into recipients of heart transplant resulted in migration of the beta-gal+ cells into the lesions of chronic rejection in the cardiac grafts and homing of the cells to the bone marrow. The majority of beta-gal+ cells present in the allografts exhibited fibroblast phenotypes, and a small number of the cells expressed desmin, indicative of myocyte differentiation. CONCLUSION: MSC vigorously migrated into the site of allograft rejection. This data suggests that they may be attracted to this site to actively participate in tissue repair during chronic rejection. In addition, given the robust migration, the inhibition of MSC differentiation toward fibroblast progeny and induction toward the myocyte lineage may serve as a new strategy for treatment of chronic rejection and allograft tissue repair.


Assuntos
Rejeição de Enxerto/fisiopatologia , Transplante de Coração , Mesoderma/citologia , Células-Tronco/fisiologia , Animais , Células Sanguíneas/patologia , Medula Óssea/patologia , Diferenciação Celular , Linhagem Celular , Movimento Celular , Transplante de Células , Doença Crônica , Colágeno , Combinação de Medicamentos , Técnicas Genéticas , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto , Óperon Lac , Laminina , Miocárdio/patologia , Reação em Cadeia da Polimerase , Proteoglicanas , Ratos , Ratos Endogâmicos Lew , Células-Tronco/patologia , Células Estromais/patologia , Células Estromais/fisiologia , Fatores de Tempo , Transplante Homólogo , Cicatrização/fisiologia
3.
Transplantation ; 76(3): 609-14, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12923453

RESUMO

BACKGROUND: Allograft fibrosis is a prominent feature of chronic rejection. Although intragraft fibroblasts contribute to this process, their origin and exact role remain poorly understood. METHODS: Using a rat model of chronic rejection, LEW to F344, cardiac fibroblasts were isolated at the point of rejection and examined in a collagen gel contraction assay to measure fibroblast activation. The allograft microenvironment was examined using immunohistochemistry for fibrogenic markers (transforming growth factor [TGF]-beta, platelet-derived growth factor [PDGF], tissue plasminogen activator [TPA], plasminogen activator inhibitor [PAI]-1, matrix metalloproteinase [MMP]-2, and tissue inhibitor of matrix metalloproteinase [TIMP]-2). The origin of intragraft fibroblasts was studied using female to male allografts followed by polymerase chain reaction [PCR] and in situ hybridization for the male sry gene. RESULTS: The cardiac fibroblasts isolated from allografts with chronic rejection exhibited higher gel contractibility (50.9% +/- 6.1% and 68.2% +/- 3.8% at 4 and 24 hr) compared with naive cardiac fibroblasts (30.7% +/- 3.5% and 55.3% +/- 6.6% at 4 and 24 hr; P<0.05 and <0.05, respectively). Immunostaining for TGF-beta, PDGF, TPA, PAI-1, MMP-2 and TIMP-2 was observed in all allografts at the time of rejection. In situ hybridization demonstrated the presence of sry positive cells in female allografts rejected by male recipients. Sixty-five percent of fibroblast colonies (55 of 85) isolated from female heart allografts expressed the male sry gene. CONCLUSION: Cardiac fibroblasts are activated and exist in a profibrogenic microenvironment in allografts undergoing chronic rejection. A substantial proportion of intragraft fibroblasts are recruited from allograft recipients in this experimental model of chronic cardiac allograft rejection.


Assuntos
Fibroblastos/fisiologia , Rejeição de Enxerto/patologia , Transplante de Coração , Animais , Doença Crônica , Feminino , Imuno-Histoquímica , Hibridização In Situ , Masculino , Metaloproteinase 2 da Matriz/análise , Inibidor 1 de Ativador de Plasminogênio/análise , Fator de Crescimento Derivado de Plaquetas/análise , Reação em Cadeia da Polimerase , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Inibidor Tecidual de Metaloproteinase-2/análise , Ativador de Plasminogênio Tecidual/análise , Fator de Crescimento Transformador beta/análise , Transplante Homólogo
4.
J Heart Lung Transplant ; 21(11): 1174-87, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12431491

RESUMO

BACKGROUND: Non-major histocompatibility complex (non-MHC) alloantibodies may play a pathogenic role in chronic rejection but remain poorly characterized. METHODS: The kinetics of alloantibody production and the mechanism by which non-MHC alloantibodies cause graft injury were investigated in a Lewis-to-Fischer 344 (LEW-to-F344) rat model of cardiac transplantation. RESULTS: Flow cytometry detected that all the F344 recipients of LEW allografts produced anti-donor immunoglobulin G (IgG) antibodies reactive with LEW lymphocytes and endothelial cells. A sub-group of recipients that rejected their grafts in 30 to 60 days exhibited markedly increased levels of anti-donor IgG antibodies (n = 6, mean fluorescence intensity [MFI]:23.85 +/- 2.7) than recipients with long-surviving allografts (n = 4, MFI:11.23 +/- 0.81; p = 0.00058). Passive transfer of anti-donor sera induced chronic rejection of LEW heart allografts in an immune non-responsiveness model of F344 rats induced by intrathymic inoculation of donor-specific lymphocytes. Immunoglobulin G antibodies purified from the anti-LEW sera exhibited complement-dependent cytotoxicity against LEW vascular endothelial cells in flow-cytometric cytotoxicity assay. The targeted endothelial cells displayed early (annexin V+) and late (TUNEL+) evidence for programmed cell death. Western blot analysis of poly (ADP-ribose) polymerase (PARP) demonstrated that the 25-kD PARP-cleavage fragment was present at the lysates of the vascular endothelial cells treated with anti-donor IgG antibodies, indicating apoptosis-associated caspase activity in these cells. In situ teminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining demonstrated that vascular endothelial cell apoptosis was consistently present in all LEW heart allografts with chronic rejection. CONCLUSIONS: Non-MHC alloantibodies are pathogenic and capable of causing chronic graft injury through an antibody-induced cell apoptosis mechanism. The results emphasize the importance of non-MHC antibodies as a common predisposing factor in the development of chronic rejection.


Assuntos
Apoptose/imunologia , Endotélio Vascular/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Isoanticorpos/sangue , Animais , Testes Imunológicos de Citotoxicidade , Endotélio Vascular/citologia , Citometria de Fluxo , Imunoglobulina G/imunologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Transplante Homólogo
5.
Stem Cells Dev ; 19(1): 47-52, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19473101

RESUMO

Pluripotency of stem cells refers to a stem cell that has the potential to differentiate into any of the three germ layers: endoderm, mesoderm, or ectoderm. Maintaining pluripotent stem cells in culture is a tedious and demanding task. Monitoring the changing pluripotency in live cells is essential for this task. Here, we report a pluripotency monitoring system in which the expression of green fluorescent protein (GFP) is under the control of the promoter of a pluripotency gene (Rex-1). The reporter system can be permanently integrated into the genome of live cells via lentiviral vectors. This pluripotency reporter system permits the long-term real-time monitoring of pluripotency changes in a live single cell and its progeny. Our data demonstrate that the BJ cell line (a normal human fibroblast cell line) that carries this hRex-GFP construct does not express GFP until it is reprogrammed to pluripotent stage. The GFP expression was progressively lost when these pluripotent hRex-GFP cells exposed to differentiation conditions. These results indicate that insertion of the hRex-GFP construct is stable in descendant cells, a finding that has particular value in tracking pluripotency of transplanted cells and their progenies in animal studies. With this hRex-GFP reporter, the pluripotency of cells can be monitored over long periods of time via the expression of GFP. Use of this reporter system will facilitate the study of stem cell pluripotency at the single-cell level, and sheds light on the molecular mechanisms of stem cell self-renewal and subsequent differentiation.


Assuntos
Genes Reporter/fisiologia , Imagem Molecular/métodos , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Sistemas Computacionais , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Regiões Promotoras Genéticas/fisiologia
6.
J Heart Lung Transplant ; 24(12): 2160-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16364866

RESUMO

BACKGROUND: Mesenchymal progenitor cells (MPC) have recently been demonstrated to actively migrate into cardiac allografts during chronic rejection. This study examines the role of MPC in tissue repair of heart allografts in a rat model of chronic rejection. METHODS: The potential of a rat MPC line (Ap8c3) to differentiate to myofibroblasts and cardiomyocytes was studied in differentiation cultures. Ap8c3 cells tagged with an enhanced green fluorescent protein (eGFP) reporter gene were engrafted into Fischer 344 (F344) recipients of Lewis (LEW) cardiac allografts. Development of intragraft MPC into scar-forming fibroblasts and cardiomyocytes was studied using immunohistochemistry. RESULTS: Ap8c3 cells contain fibroblast progenitors (FP) positive for P07 antibody. Transforming growth factor (TGF)-beta stimulation promoted FP to terminally differentiate into myofibroblasts, which express alpha-smooth muscle actin (alphaSMA). In cardiac differentiation culture, Ap8c3 cells were induced by 5-azatiditin (5-aza) to form tropomyosin+ myotubes, and to express mRNA encoding for cardiac troponin I (TnI) and alpha-myosin heavy chain (alphaMHC). Transfusion of eGFP+ Ap8c3 cells to F344 recipients resulted in migration of eGFP(+) cells into LEW heart allografts, as well as homing of the eGFP+ MPC to bone marrow. The majority of eGFP+ cells in the heart allografts appeared to be vimentin-expressing fibroblasts. Foci of eGFP+ myocardium were also detected in all heart allografts, with eGFP+ cardiomyocytes representing 4.8 +/- 1.2% of the allografted eGFP+ cells. CONCLUSIONS: The data suggest that rat MPC participate in tissue repair in heart allografts by giving rise to scar-forming myofibroblasts and cardiomyocytes.


Assuntos
Rejeição de Enxerto/terapia , Transplante de Coração/imunologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Regeneração , Animais , Diferenciação Celular , Movimento Celular , Doença Crônica , Fibroblastos , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Miócitos Cardíacos , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Transplante Heterotópico , Transplante Homólogo
7.
Immunology ; 112(1): 87-93, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15096188

RESUMO

CXCR3 chemokines are of particular interest because of their potential involvement in a variety of inflammatory diseases, including the rejection of organ transplants. Although the rat is one of the most appropriate animals for using to study transplantation biology, the structural and functional characteristics of CXCL9 [monokine induced by interferon-gamma (Mig)] in this experimental model have not been described. Therefore, we recently conducted a series of experiments to identify and characterize the rat CXCL9 gene. Accordingly, we isolated rat CXCL9 cDNA and genomic DNA. The rat CXCL9 gene encodes a protein of 125 amino acids and spans a 3.5 kbp DNA segment containing four exons in the protein-coding region. We then analysed mRNA expression in various tissues. Transcripts for the gene were found to be expressed at high levels in the lymph nodes and spleen. Then, to confirm the function of the identified gene, rat CXCL9 was transiently expressed in COS-1 cells. Rat recombinant Mig displayed chemotactic properties and induced CXCR3 internalization in CD4+ T cells. Lastly, we analysed the expression of rat CXCL9 in a heterotopic heart allograft model. Both mRNA and protein levels of intragraft CXCL9 were significantly increased following transplantation of ACI to LEW hearts when compared with syngeneic controls. These findings indicate that rat CXCL9 has an in vivo role in the infiltration of CD4+ T cells in the transplanted graft.


Assuntos
Quimiocinas CXC/genética , Transplante de Coração/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Sequência de Aminoácidos , Animais , Anti-Inflamatórios/farmacologia , Células COS , Quimiocina CXCL9 , Quimiocinas CXC/imunologia , DNA Complementar/genética , Dexametasona/farmacologia , Expressão Gênica , Glucocorticoides/farmacologia , Sobrevivência de Enxerto/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Interferon gama/antagonistas & inibidores , Interferon gama/farmacologia , Linfonodos/imunologia , Masculino , Dados de Sequência Molecular , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos ACI , Proteínas Recombinantes , Baço/imunologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA