Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Proteome Res ; 13(11): 4901-9, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25300029

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disorder caused by mutations in the Pkd1 or Pkd2 genes, in which large cysts replace normal kidney tissue, leading to end-stage kidney disease. In this study we have utilized a powerful nano-HPLC-mass spectrometric approach to characterize patterns of normal and abnormal N-linked glycosylation of α3 integrin subunit in Pkd1(-/-) cells derived from mouse kidneys. Higher molecular weight glycan structures with a different monosaccharide composition were observed at two sites, namely, Asn-925 and Asn-928 sites in α3 integrin isolated from Pkd1(+/+) cells compared with Pkd1(-/-) cells. In addition, an unusual and unique disialic acid glycan structure was observed solely in Pkd1(-/-) cells. Thus, these studies suggest that abnormal protein glycosylation may have a role on the pathogenesis of cyst formation in ADPKD.


Assuntos
Integrina alfa3/metabolismo , Doenças Renais Policísticas/metabolismo , Polissacarídeos/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Imunoprecipitação , Espectrometria de Massas , Camundongos , Camundongos Knockout , Doenças Renais Policísticas/patologia , Polissacarídeos/isolamento & purificação , Ácidos Siálicos/metabolismo , Canais de Cátion TRPP/genética
2.
Stem Cells ; 31(10): 2095-103, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23836547

RESUMO

Human embryonic stem cells (hESCs) can be progressively differentiated into definitive endoderm (DE), hepatic progenitors, and hepatocytes, and thus provide an excellent model system for the mechanistic study of hepatocyte differentiation, which is currently poorly understood. Here, we found that insulin enhanced hepatocyte differentiation from hESC-derived DE. Insulin activated the PI3K/AKT pathway, but not the mitogen-activated protein kinase pathway in the DE cells, and inhibition of the PI3K/AKT pathways by inhibitors markedly inhibited hepatocyte differentiation. In addition, insulin-like growth factor 1 (IGF1) and IGF2 also activated the PI3K/AKT pathway in DE cells and their expression was robustly upregulated during hepatocyte differentiation from DE. Furthermore, inhibition of IGF receptor 1 (IGF1R) by a small molecule inhibitor PPP or knockdown of the IGF1R by shRNA attenuated hepatocyte differentiation. Moreover, simultaneous knockdown of the IGF1R and the insulin receptor with shRNAs markedly reduced the activation of AKT and substantially impaired hepatocyte differentiation. The PI3K pathway specifically enhanced the expression of HNF1 and HNF4 to regulate hepatocyte differentiation from DE. Although inhibition of the PI3K pathway was previously shown to be required for the induction of DE from hESCs, our study revealed a positive role of the PI3K pathway in hepatocyte differentiation after the DE stage, and has advanced our understanding of hepatocyte cell fate determination.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Hepatócitos/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Insulina/fisiologia , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Linhagem Celular , Sobrevivência Celular , Humanos , Fator de Crescimento Insulin-Like II/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor IGF Tipo 1/fisiologia , Receptor de Insulina/fisiologia , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma
3.
Stem Cells ; 30(1): 42-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21898694

RESUMO

Adult stem cell therapies have provided success for more than 50 years, through reconstitution of the hematopoietic system using bone marrow, umbilical cord blood, and mobilized peripheral blood transplantation. Mesenchymal stem cell (MSC)-mediated therapy is a fast-growing field that has proven safe and effective in the treatment of various degenerative diseases and tissue injuries. Since the first derivation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), there has been impressive progress toward developing safe clinical applications from PSCs. Recent successes in transgene-free iPSC reprogramming have brought attention to the potential of clinical applications of these pluripotent cells, but key hurdles must be overcome, which are discussed in this review. Looking to the future, it could be advantageous to derive MSC from iPSC or human ESC in cases where genetic engineering is needed, since in the PSCs, clones with "safe harbor" vector integration could be selected, expanded, and differentiated. Here, we describe the status of the progress of the use of MSC and PSCs in clinical trials and analyze the challenges that should be overcome before iPSC-derived MSC therapy can be used widely in the clinic.


Assuntos
Células-Tronco Adultas/transplante , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Adultas/metabolismo , Animais , Diferenciação Celular/fisiologia , Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco , Transgenes/genética
4.
Mol Ther ; 19(3): 584-93, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21119622

RESUMO

Induced pluripotent stem cells (iPSCs) have radically advanced the field of regenerative medicine by making possible the production of patient-specific pluripotent stem cells from adult individuals. By developing iPSCs to treat HIV, there is the potential for generating a continuous supply of therapeutic cells for transplantation into HIV-infected patients. In this study, we have used human hematopoietic stem cells (HSCs) to generate anti-HIV gene expressing iPSCs for HIV gene therapy. HSCs were dedifferentiated into continuously growing iPSC lines with four reprogramming factors and a combination anti-HIV lentiviral vector containing a CCR5 short hairpin RNA (shRNA) and a human/rhesus chimeric TRIM5α gene. Upon directed differentiation of the anti-HIV iPSCs toward the hematopoietic lineage, a robust quantity of colony-forming CD133(+) HSCs were obtained. These cells were further differentiated into functional end-stage macrophages which displayed a normal phenotypic profile. Upon viral challenge, the anti-HIV iPSC-derived macrophages exhibited strong protection from HIV-1 infection. Here, we demonstrate the ability of iPSCs to develop into HIV-1 resistant immune cells and highlight the potential use of iPSCs for HIV gene and cellular therapies.


Assuntos
Infecções por HIV/imunologia , HIV-1/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Antígeno AC133 , Adulto , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Glicoproteínas/metabolismo , Células HEK293 , Infecções por HIV/virologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Peptídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores CCR5/genética , Receptores CCR5/metabolismo
5.
Stem Cells Dev ; 17(2): 315-24, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18447646

RESUMO

Self-renewing pluripotent human embryonic stem (hES) cells are capable of regenerating such non-dividing cells as neurons and cardiomyocytes for therapies and can serve as an excellent experimental model for studying early human development. Both the spatial and temporal relationships of gene expression play a crucial role in determining differentiation; to obtain a better understanding of hES cell differentiation, it will be necessary to establish an inducible system in hES cells that enables specific transgene(s) to reversibly and conditionally express (1) at specific levels and (2) at particular time points during development. Using lentivirus (LV)-mediated gene transfer and a tetracycline-controlled trans-repressor (TR), we first established in hES cells a doxycycline (DOX)-inducible expression system of green fluorescent protein (GFP) to probe its reversibility and kinetics. Upon the addition of DOX, the percentage of GFP(+) hES cells increased time dependently: The time at which 50% of all green cells appeared (T(50)(on)) was 119.5+/-3.2 h; upon DOX removal, GFP expression declined with a half-time (T(50)(off)) of 127.7+/-3.9 h and became completely silenced at day 8. Both the proportion and total mean fluorescence intensity (MFI) were dose-dependent (EC(50)=24.5+/-2.2 ng/ml). The same system when incorporated into murine (m) ES cells similarly exhibited reversible dose-dependent responses with a similar sensitivity (EC(50)=49.5+/-8.5 ng/ml), but the much faster kinetics (T(50)(on)=35.5+/-5.5 h, T(50)(off) = 71.5+/-2.4 hours). DOX-induced expression of the Kir2.1 channels in mES and hES cells led to robust expression of the inwardly rectifying potassium (K(+)) current and thereby hyperpolarized the resting membrane potential (RMP). We conclude that the LV-inducible system established presents a unique tool for probing differentiation.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica , Transgenes , Animais , Células Cultivadas , Clonagem Molecular , Relação Dose-Resposta a Droga , Doxiciclina/farmacologia , Eletrofisiologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Potenciais da Membrana/genética , Camundongos , Modelos Biológicos , Fenótipo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
6.
Curr Stem Cell Res Ther ; 11(2): 131-40, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26763885

RESUMO

We have previously described generation of mesenchymal stem cells (MSCs) from human embryonic and induced pluripotent stem cells. One of the central questions in stem cell biology is to understand how stem cells regulate the decision to self-renew vs. differentiate, at the molecular level. In the current studies we used loss-of-function and gain-of-function analyses in primary human MSCs to demonstrate that BMI1 is a critical regulator for self-renewal and multipotency in this interesting cell type. Knockdown of BMI1 in MSCs reduced self-renewal by upregulation of p16(INK4A) and increased apoptosis. Knockdown of p16(INK4A) partially rescued the self-renewal defect in MSCs with loss of BMI1. Overexpressed BMI1 reduced apoptosis and increased cell proliferation by repressing p16(INK4A). Loss of BMI1 resulted in deregulation of PPARγ, an adipogenic factor, and imprinted gene network (IGN), which blocks osteogenesis. Knockdown of PPARγ or IGN in BMI1 defect models restored osteogenesis. Overexpression of BMI1 repressed transcripts of RUNX2 and PPARγ, in osteogenesis and adipogenesis, respectively, which lead to decreased lineage specification potential in MSCs. These data show that BMI1 regulates cell proliferation, apoptosis, and differentiation of human MSCs.


Assuntos
Diferenciação Celular/genética , Células-Tronco Mesenquimais/citologia , Proteína Quinase 7 Ativada por Mitógeno/genética , Osteogênese/genética , Apoptose/genética , Linhagem da Célula , Proliferação de Células/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , PPAR gama/genética
7.
Curr Top Dev Biol ; 107: 405-38, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24439814

RESUMO

Tissue homeostasis and regenerative capacity rely on rare populations of somatic stem cells endowed with the potential to self-renew and differentiate. During aging, many tissues show a decline in regenerative potential coupled with a loss of stem cell function. Cells including somatic stem cells have evolved a series of checks and balances to sense and repair cellular damage to maximize tissue function. However, during aging the mechanisms that protect normal cell function begin to fail. In this review, we will discuss how common cellular mechanisms that maintain tissue fidelity and organismal lifespan impact somatic stem cell function. We will highlight context-dependent changes and commonalities that define aging, by focusing on three age-sensitive stem cell compartments: blood, neural, and muscle. Understanding the interaction between extrinsic regulators and intrinsic effectors that operate within different stem cell compartments is likely to have important implications for identifying strategies to improve health span and treat age-related degenerative diseases.


Assuntos
Células-Tronco Adultas/fisiologia , Envelhecimento/fisiologia , Microambiente Celular , Homeostase/fisiologia , Modelos Biológicos , Regeneração/fisiologia , Células-Tronco Adultas/citologia , Células Sanguíneas/citologia , DNA Mitocondrial/genética , Metabolismo Energético/fisiologia , Epigênese Genética/genética , Epigênese Genética/fisiologia , Instabilidade Genômica/fisiologia , Humanos , Músculos/citologia , Tecido Nervoso/citologia
8.
Adv Drug Deliv Rev ; 62(12): 1167-74, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20920540

RESUMO

Mesenchymal stem cells (MSC) are a promising tool for cell therapy, either through direct contribution to the repair of bone, tendon and cartilage or as an adjunct therapy through protein production and immune mediation. They are an attractive vehicle for cellular therapies due to a variety of cell intrinsic and environmentally responsive properties. Following transplantation, MSC are capable of systemic migration, are not prone to tumor formation, and appear to tolerize the immune response across donor mismatch. These attributes combine to allow MSC to reside in many different tissue types without disrupting the local microenvironment and, in some cases, responding to the local environment with appropriate protein secretion. We describe work done by our group and others in using human MSC for the sustained in vivo production of supraphysiological levels of cytokines for the support of cotransplanted hematopoietic stem cells and enzymes that are deficient in animal models of lysosomal storage disorders such as MPSVII. In addition, the use of MSC engineered to secrete protein products has been reviewed in several fields of tissue injury repair, including but not limited to revascularization after myocardial infarction, regeneration of intervertebral disc defects and spine therapy, repair of stroke, therapy for epilepsy, skeletal tissue repair, chondrogenesis/knee and joint repair, and neurodegenerative diseases. Genetically engineered MSC have thus proven safe and efficacious in numerous animal models of disease modification and tissue repair and are poised to be tested in human clinical trials. The potential for these interesting cells to secrete endogenous or transgene products in a sustained and long-term manner is highly promising and is discussed in the current review.


Assuntos
Citocinas/metabolismo , Sistemas de Liberação de Medicamentos , Engenharia Genética , Fatores Imunológicos/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ensaios Clínicos como Assunto , Citocinas/genética , Transplante de Células-Tronco Hematopoéticas , Humanos , Fatores Imunológicos/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/metabolismo , Mucopolissacaridose VII/terapia , Regeneração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA