Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuro Oncol ; 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38982561

RESUMO

BACKGROUND: Chimeric antigen receptor (CAR)-T cell therapies targeting glioblastoma (GBM)-associated antigens such as interleukin-13 receptor subunit alpha-2 (IL-13Rα2) have achieved limited clinical efficacy to date, in part due to an immunosuppressive tumor microenvironment (TME) characterized by inhibitory molecules such as transforming growth factor-beta (TGF-ß). The aim of this study was to engineer more potent GBM-targeting CAR-T cells by countering TGF-ß-mediated immune suppression in the TME. METHODS: We engineered a single-chain, bispecific CAR targeting IL-13Rα2 and TGF-ß, which programs tumor-specific T cells to convert TGF-ß from an immunosuppressant to an immunostimulant. Bispecific IL-13Rα2/TGF-ß CAR-T cells were evaluated for efficacy and safety against both patient-derived GBM xenografts and syngeneic models of murine glioma. RESULTS: Treatment with IL-13Rα2/TGF-ß CAR-T cells leads to greater T-cell infiltration and reduced suppressive myeloid cell presence in the tumor-bearing brain compared to treatment with conventional IL-13Rα2 CAR-T cells, resulting in improved survival in both patient-derived GBM xenografts and syngeneic models of murine glioma. CONCLUSION: Our findings demonstrate that by reprogramming tumor-specific T-cell responses to TGF-ß, bispecific IL-13Rα2/TGF-ß CAR-T cells resist and remodel the immunosuppressive TME to drive potent anti-tumor responses in GBM.

2.
J Vasc Interv Radiol ; 35(6): 809-817.e1, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38219903

RESUMO

Traditionally, rodent cancer models have driven preclinical oncology research. However, they do not fully recapitulate characteristics of human cancers, and their size poses challenges when evaluating tools in the interventional oncologists' armamentarium. Pig models, however, have been the gold standard for validating surgical procedures. Their size enables the study of image-guided interventions using human ultrasound (US), computed tomography (CT), and magnetic resonance (MR) imaging platforms. Furthermore, pigs have immunologic features that are similar to those of humans, which can potentially be leveraged for studying immunotherapy. Novel pig models of cancer are being developed, but additional research is required to better understand both the pig immune system and malignancy to enhance the potential for pig models in interventional oncology research. This review aims to address the main advantages and disadvantages of using a pig model for interventional oncology and outline the specific characteristics of pig models that make them more suitable for investigation of locoregional therapies.


Assuntos
Modelos Animais de Doenças , Imunoterapia , Neoplasias , Animais , Imunoterapia/métodos , Neoplasias/terapia , Neoplasias/diagnóstico por imagem , Neoplasias/imunologia , Humanos , Suínos , Radiografia Intervencionista , Sus scrofa , Oncologia
3.
Cell Stem Cell ; 25(4): 542-557.e9, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31495780

RESUMO

Invariant natural killer T (iNKT) cells are potent immune cells for targeting cancer; however, their clinical application has been hindered by their low numbers in cancer patients. Here, we developed a proof-of-concept for hematopoietic stem cell-engineered iNKT (HSC-iNKT) cell therapy with the potential to provide therapeutic levels of iNKT cells for a patient's lifetime. Using a human HSC engrafted mouse model and a human iNKT TCR gene engineering approach, we demonstrated the efficient and long-term generation of HSC-iNKT cells in vivo. These HSC-iNKT cells closely resembled endogenous human iNKT cells, could deploy multiple mechanisms to attack tumor cells, and effectively suppressed tumor growth in vivo in multiple human tumor xenograft mouse models. Preclinical safety studies showed no toxicity or tumorigenicity of the HSC-iNKT cell therapy. Collectively, these results demonstrated the feasibility, safety, and cancer therapy potential of the proposed HSC-iNKT cell therapy and laid a foundation for future clinical development.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Imunoterapia Adotiva/métodos , Células T Matadoras Naturais/fisiologia , Neoplasias/terapia , Animais , Células Cultivadas , Engenharia Genética , Humanos , Camundongos , Camundongos SCID , Células T Matadoras Naturais/transplante , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nat Cell Biol ; 18(6): 595-606, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27183470

RESUMO

Pluripotent stem cells (PSCs) may provide a potential source of haematopoietic stem/progenitor cells (HSPCs) for transplantation; however, unknown molecular barriers prevent the self-renewal of PSC-HSPCs. Using two-step differentiation, human embryonic stem cells (hESCs) differentiated in vitro into multipotent haematopoietic cells that had the CD34(+)CD38(-/lo)CD90(+)CD45(+)GPI-80(+) fetal liver (FL) HSPC immunophenotype, but exhibited poor expansion potential and engraftment ability. Transcriptome analysis of immunophenotypic hESC-HSPCs revealed that, despite their molecular resemblance to FL-HSPCs, medial HOXA genes remained suppressed. Knockdown of HOXA7 disrupted FL-HSPC function and caused transcriptome dysregulation that resembled hESC-derived progenitors. Overexpression of medial HOXA genes prolonged FL-HSPC maintenance but was insufficient to confer self-renewal to hESC-HSPCs. Stimulation of retinoic acid signalling during endothelial-to-haematopoietic transition induced the HOXA cluster and other HSC/definitive haemogenic endothelium genes, and prolonged HSPC maintenance in culture. Thus, medial HOXA gene expression induced by retinoic acid signalling marks the establishment of the definitive HSPC fate and controls HSPC identity and function.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula , Genes Homeobox/genética , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Multipotentes/citologia , Antígenos CD34/metabolismo , Células Cultivadas , Perfilação da Expressão Gênica/métodos , Proteínas de Homeodomínio/genética , Humanos , Antígenos Comuns de Leucócito/metabolismo , Transcriptoma
5.
J Enzyme Inhib Med Chem ; 31(6): 999-1004, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26307919

RESUMO

Recently it was found that dipotassium-trioxohydroxytetrafluorotriborate, K2(B3O3F4OH), is a potent and highly specific inhibitor of precancerous cell processes. We conducted gene expression profiling of human melanoma cells before and after treatment with two concentrations (0.1 and 1 mM) of this boron inorganic derivative in order to assess its effects on deregulation of genes associated with tumor pathways. Parallel trypan blue exclusion assay was performed to assess the cytotoxicity effects of this chemical. Treatment with K2(B3O3F4OH) induced a significant decrease of cell viability in melanoma cellline at both tested concentrations. Furthermore, these treatments caused deregulation of more than 30 genes known as common anti-tumor drug targets. IGF-1 and hTERT were found to be significantly downregulated and this result may imply potential use of K2(B3O3F4OH) as an inhibitor or human telomerase and insulin-like growth factor 1, both of which are associated with various tumor pathways.


Assuntos
Antineoplásicos/farmacologia , Compostos de Boro/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/genética , Melanoma/tratamento farmacológico , Melanoma/genética , Telomerase/genética , Antineoplásicos/síntese química , Antineoplásicos/química , Compostos de Boro/síntese química , Compostos de Boro/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Estrutura Molecular , Relação Estrutura-Atividade , Telomerase/metabolismo
6.
Cell Stem Cell ; 16(4): 367-72, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25772073

RESUMO

X-linked Severe Combined Immunodeficiency (SCID-X1) is a genetic disease that leaves newborns at high risk of serious infection and a predicted life span of less than 1 year in the absence of a matched bone marrow donor. The disease pathogenesis is due to mutations in the gene encoding the Interleukin-2 receptor gamma chain (IL-2Rγ), leading to a lack of functional lymphocytes. With the leukemogenic concerns of viral gene therapy there is a need to explore alternative therapeutic options. We have utilized induced pluripotent stem cell (iPSC) technology and genome editing mediated by TALENs to generate isogenic subject-specific mutant and gene-corrected iPSC lines. While the subject-derived mutant iPSCs have the capacity to generate hematopoietic precursors and myeloid cells, only wild-type and gene-corrected iPSCs can additionally generate mature NK cells and T cell precursors expressing the correctly spliced IL-2Rγ. This study highlights the potential for the development of autologous cell therapy for SCID-X1 subjects.


Assuntos
Terapia Genética/métodos , Imunoterapia Adotiva , Células-Tronco Pluripotentes Induzidas/fisiologia , Células Matadoras Naturais/fisiologia , Células Precursoras de Linfócitos T/fisiologia , Regeneração , Medicina Regenerativa , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/terapia , Antígenos CD/metabolismo , Proteínas de Bactérias/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Lactente , Subunidade gama Comum de Receptores de Interleucina/genética , Células Matadoras Naturais/transplante , Mutação/genética , Células Precursoras de Linfócitos T/transplante , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética
7.
J Enzyme Inhib Med Chem ; 30(3): 354-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24939106

RESUMO

Dipotassium-trioxohydroxytetrafluorotriborate K2[B3O3F4OH] was listed as a promising new therapeutic for cancer diseases. For in vitro and in vivo investigation of its antitumor effects 4T1 mammary adenocarcinoma, B16F10 melanoma and squamous cell carcinoma SCCVII were used. The detailed in vitro investigation undoubtedly showed that K2[B3O3F4OH] affects the growth of cancer cells. The proliferation of cells depends on the concentration so that aqueous solution of K2[B3O3F4OH], the concentrations of 10(-4) M and less, does not affect cell growth, but the concentrations of 10(-3) M or more, significantly slows cells growth. B16F10 and SCCVII cells show higher sensitivity to the cytotoxic effects of K2[B3O3F4OH] compared to 4T1 cells. Under in vivo conditions, K2[B3O3F4OH] slows the growth of all three tumors tested compared to the control, and the inhibitory effect was most pronounced during the application of the substance. There is almost no difference if K2[B3O3F4OH] was applied intraperitoneally, intratumor, peroral or as ointment. Addition of 5-FU did not further increase the antitumor efficacy of K2[B3O3F4OH].


Assuntos
Antineoplásicos/farmacologia , Compostos de Boro/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Compostos de Boro/síntese química , Compostos de Boro/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Halogenação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Estrutura Molecular , Relação Estrutura-Atividade
8.
Proc Natl Acad Sci U S A ; 108(51): E1408-16, 2011 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-22123951

RESUMO

The goal of cancer immunotherapy is the generation of an effective, stable, and self-renewing antitumor T-cell population. One such approach involves the use of high-affinity cancer-specific T-cell receptors in gene-therapy protocols. Here, we present the generation of functional tumor-specific human T cells in vivo from genetically modified human hematopoietic stem cells (hHSC) using a human/mouse chimera model. Transduced hHSC expressing an HLA-A*0201-restricted melanoma-specific T-cell receptor were introduced into humanized mice, resulting in the generation of a sizeable melanoma-specific naïve CD8(+) T-cell population. Following tumor challenge, these transgenic CD8(+) T cells, in the absence of additional manipulation, limited and cleared human melanoma tumors in vivo. Furthermore, the genetically enhanced T cells underwent proper thymic selection, because we did not observe any responses against non-HLA-matched tumors, and no killing of any kind occurred in the absence of a human thymus. Finally, the transduced hHSC established long-term bone marrow engraftment. These studies present a potential therapeutic approach and an important tool to understand better and to optimize the human immune response to melanoma and, potentially, to other types of cancer.


Assuntos
Antineoplásicos/farmacologia , Linfócitos T CD8-Positivos/citologia , Células-Tronco Hematopoéticas/citologia , Animais , Antígenos CD34/biossíntese , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Citometria de Fluxo/métodos , Engenharia Genética/métodos , Humanos , Imunoterapia/métodos , Linfócitos do Interstício Tumoral/citologia , Camundongos , Camundongos SCID , Modelos Genéticos , Transplante de Neoplasias , Células-Tronco/citologia , Timo/metabolismo , Transgenes
9.
Blood ; 115(8): 1534-44, 2010 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-20018916

RESUMO

Inhibiting the expression of the HIV-1 coreceptor CCR5 holds great promise for controlling HIV-1 infection in patients. Here we report stable knockdown of human CCR5 by a short hairpin RNA (shRNA) in a humanized bone marrow/liver/thymus (BLT) mouse model. We delivered a potent shRNA against CCR5 into human fetal liver-derived CD34(+) hematopoietic progenitor/stem cells (HPSCs) by lentiviral vector transduction. We transplanted vector-transduced HPSCs solidified with Matrigel and a thymus segment under the mouse kidney capsule. Vector-transduced autologous CD34(+) cells were subsequently injected in the irradiated mouse, intended to create systemic reconstitution. CCR5 expression was down-regulated in human T cells and monocytes/macrophages in systemic lymphoid tissues, including gut-associated lymphoid tissue, the major site of HIV-1 replication. The shRNA-mediated CCR5 knockdown had no apparent adverse effects on T-cell development as assessed by polyclonal T-cell receptor Vbeta family development and naive/memory T-cell differentiation. CCR5 knockdown in the secondary transplanted mice suggested the potential of long-term hematopoietic reconstitution by the shRNA-transduced HPSCs. CCR5 tropic HIV-1 infection was effectively inhibited in mouse-derived human splenocytes ex vivo. These results demonstrate that lentiviral vector delivery of shRNA into human HPSCs could stably down-regulate CCR5 in systemic lymphoid organs in vivo.


Assuntos
Medula Óssea/metabolismo , Infecções por HIV/metabolismo , HIV-1 , Transplante de Células-Tronco Hematopoéticas , Fígado/metabolismo , Receptores CCR5/biossíntese , Timo/metabolismo , Animais , Diferenciação Celular/genética , Modelos Animais de Doenças , Regulação para Baixo , Técnicas de Silenciamento de Genes , Infecções por HIV/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Memória Imunológica/genética , Lentivirus , Camundongos , Camundongos Endogâmicos NOD , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores CCR5/genética , Linfócitos T/metabolismo , Transdução Genética , Transplante Heterólogo
10.
PLoS One ; 4(12): e8208, 2009 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-19997617

RESUMO

There is a desperate need for effective therapies to fight chronic viral infections. The immune response is normally fastidious at controlling the majority of viral infections and a therapeutic strategy aimed at reestablishing immune control represents a potentially powerful approach towards treating persistent viral infections. We examined the potential of genetically programming human hematopoietic stem cells to generate mature CD8+ cytotoxic T lymphocytes that express a molecularly cloned, "transgenic" human anti-HIV T cell receptor (TCR). Anti-HIV TCR transduction of human hematopoietic stem cells directed the maturation of a large population of polyfunctional, HIV-specific CD8+ cells capable of recognizing and killing viral antigen-presenting cells. Thus, through this proof-of-concept we propose that genetic engineering of human hematopoietic stem cells will allow the tailoring of effector T cell responses to fight HIV infection or other diseases that are characterized by the loss of immune control.


Assuntos
Antígenos Virais/imunologia , Epitopos/imunologia , Engenharia Genética , Células-Tronco Hematopoéticas/imunologia , Linfócitos T/imunologia , Animais , Antivirais/metabolismo , Diferenciação Celular/imunologia , Clonagem Molecular , HIV/imunologia , Humanos , Camundongos , Camundongos SCID , Fenótipo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/isolamento & purificação , Especificidade da Espécie , Linfócitos T/virologia
11.
Stem Cells ; 27(1): 18-28, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19224508

RESUMO

Human germ cell tumors are often metastatic, presumably due to distal site tumor growth by cancer stem cells. To determine whether cancer stem cells can be identified in a transplantation model of testicular germ cell tumor, we transplanted murine embryonic germ cells (EGCs) into the testis of adult severe combined immunodeficient mice. Transplantation resulted in a locally invasive solid tumor, with a cellular component that generated secondary tumors upon serial transplantation. The secondary tumors were invariably metastatic, a feature not observed in the primary tumors derived from EGCs. To characterize the differences between EGCs and the tumor-derived stem cells, we performed karyotype and microarray analysis. Our results show that generation of cancer stem cells is associated with the acquisition of nonclonal genomic rearrangements not found in the originating population. Furthermore, pretreatment of EGCs with a potent inhibitor of self-renewal, retinoic acid, prevented tumor formation and the emergence of these genetically unstable cancer stem cells. Microarray analysis revealed that EGCs and first- and second-generation cancer stem cells were highly similar; however, approximately 1,000 differentially expressed transcripts could be identified corresponding to alterations in oncogenes and genes associated with motility and development. Combined, the data suggest that the activation of oncogenic pathways in a cellular background of genetic instability, coupled with an inherent ability to self-renew, is involved in the acquisition of metastatic behavior in the cancer stem cell population of tumors derived from pluripotent cells.


Assuntos
Instabilidade Genômica , Células-Tronco Neoplásicas/patologia , Células-Tronco Pluripotentes/patologia , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Imunofluorescência , Instabilidade Genômica/efeitos dos fármacos , Células Germinativas/efeitos dos fármacos , Células Germinativas/transplante , Humanos , Antígenos CD15/metabolismo , Masculino , Camundongos , Modelos Biológicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco Pluripotentes/efeitos dos fármacos , Neoplasias Testiculares/patologia , Tretinoína/farmacologia
12.
Stem Cells ; 27(1): 100-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18974209

RESUMO

Human embryonic stem cells (hESC) have the potential to revolutionize certain medical treatments, including T-cell-based therapies. However, optimal approaches to develop T cells from hESC are lacking. In this report, we show that T-cell progenitors can be derived from hESC cultured as embryoid bodies (EBs). These EB-derived T-cell progenitors give rise to phenotypically and functionally normal cells of the T lineage when transferred into human thymic tissue implanted in immunocompromised mice, suggesting that introduction of these progenitors into patients may also yield functional T cells. Moreover, hematopoietic progenitors demonstrating T-cell potential appeared to be CD45+/CD34+, resembling those found in normal bone marrow. In contrast to T cells developed from hESC cocultured on murine stromal cells, the EB-derived T cells also expressed normal levels of CD45. Importantly, the EB system eliminates the previous need for murine cocultures, a key impediment to developing a protocol for T-cell progenitor derivation suitable for clinical use. Furthermore, following lentiviral-mediated introduction of a vector expressing enhanced green fluorescent protein into hESC, stable transgene expression was maintained throughout differentiation, suggesting a potential for gene therapy approaches aimed at the augmentation of T-cell function or treatment of T-cell disorders.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias/citologia , Linfócitos T/citologia , Animais , Diferenciação Celular , Linhagem Celular , Embrião de Mamíferos/citologia , Proteínas de Fluorescência Verde/metabolismo , Hematopoese , Humanos , Cinética , Camundongos , Fenótipo
13.
J Stem Cells ; 4(1): 29-45, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20498689

RESUMO

Human embryonic stem cells can differentiate into CD34+ hematopoietic progenitors by co-culture on murine feeders such as OP9 and S17. These CD34+ progenitors can be further differentiated into several cells of the hematopoietic lineage including macrophages. However, co-culture on murine feeders is time consuming and involves extensive manipulations. Furthermore, CD45 expression is low on hematopoietic cultures derived from stromal co-cultures. In this study we describe a novel and highly efficient system of generating differentiated macrophages from hematopoietic progenitors generated from embryoid body cultures of human embryonic stem cells. The hematopoietic progenitors generated from these embryoid bodies express higher numbers of CD45+ cells and are able to differentiate to macrophages when cultured in presence of cytokines. Using this system we were able to generate higher yields of CD14+ macrophages compared to traditional stromal cell culture methods. The embryoid body derived macrophages are phagocytic, respond to Toll-like receptor stimulation and express phenotypic markers of mature macrophages. Importantly, the embryoid body system generates hematopoietic progenitors suitable for clinical use by eliminating the need for murine feeder cells. Furthermore, this system is amenable to genetic manipulation and may thus be used to study important mechanisms of macrophage differentiation and function.


Assuntos
Corpos Embrioides , Células-Tronco Hematopoéticas , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Embrionárias Humanas , Humanos , Macrófagos
14.
Genes Dev ; 22(22): 3121-34, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19056892

RESUMO

Loss of the CDK inhibitor p27(KIP1) is widely linked with poor prognosis in human cancer. In Wnt10b-expressing mammary tumors, levels of p27(KIP1) were extremely low; conversely, Wnt10b-null mammary cells expressed high levels of this protein, suggesting Wnt-dependent regulation of p27(KIP1). Interestingly we found that Wnt-induced turnover of p27(KIP1) was independent from classical SCF(SKP2)-mediated degradation in both mouse and human cells. Instead, turnover required Cullin 4A and Cullin 4B, components of an alternative E3 ubiquitin ligase induced in response to active Wnt signaling. We found that CUL4A was a novel Wnt target gene in both mouse and human cells and that CUL4A physically interacted with p27(KIP1) in Wnt-responding cells. We further demonstrated that both Cul4A and Cul4B were required for Wnt-induced p27(KIP1) degradation and S-phase progression. CUL4A and CUL4B are therefore components of a conserved Wnt-induced proteasome targeting (WIPT) complex that regulates p27(KIP1) levels and cell cycle progression in mammalian cells.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteínas Culina/genética , Proteínas Culina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Feminino , Humanos , Immunoblotting , Imunoprecipitação , Carioferinas/genética , Carioferinas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Transdução de Sinais/genética , Proteínas Wnt/genética , Proteína Exportina 1
15.
Proc Natl Acad Sci U S A ; 103(31): 11742-7, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16844782

RESUMO

Harnessing the ability of genetically manipulated human embryonic stem cells (hESC) to differentiate into appropriate lineages could revolutionize medical practice. These cells have the theoretical potential to develop into all mature cell types; however, the actual ability to develop into all hematopoietic lineages has not been demonstrated. Using sequential in vitro coculture on murine bone marrow stromal cells, and engraftment into human thymic tissues in immunodeficient mice, we demonstrate that hESC can differentiate through the T lymphoid lineage. Stable transgene expression was maintained at high levels throughout differentiation, suggesting that genetically manipulated hESC hold potential to treat several T cell disorders.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula , Embrião de Mamíferos/citologia , Células-Tronco/fisiologia , Linfócitos T/fisiologia , Animais , Antígenos CD/metabolismo , Células Cultivadas , Técnicas de Cocultura , Humanos , Camundongos , Camundongos SCID , Transplante de Células-Tronco , Células-Tronco/citologia , Células Estromais/citologia , Células Estromais/fisiologia , Linfócitos T/citologia , Transplante de Tecidos
16.
J Virol ; 79(21): 13538-47, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16227274

RESUMO

Reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV) lytic replication is mediated by the viral RTA transcription factor, but little is known about the physiological processes controlling its expression or activity. Links between autonomic nervous system activity and AIDS-associated Kaposi's sarcoma led us to examine the potential influence of catecholamine neurotransmitters. Physiological concentrations of epinephrine and norepinephrine efficiently reactivated lytic replication of KSHV in latently infected primary effusion lymphoma cells via beta-adrenergic activation of the cellular cyclic AMP/protein kinase A (PKA) signaling pathway. Effects were blocked by PKA antagonists and mimicked by pharmacological and physiological PKA activators (prostaglandin E2 and histamine) or overexpression of the PKA catalytic subunit. PKA up-regulated RTA gene expression, enhanced activity of the RTA promoter, and posttranslationally enhanced RTA's trans-activating capacity for its own promoter and heterologous lytic promoters (e.g., the viral PAN gene). Mutation of predicted phosphorylation targets at RTA serines 525 and 526 inhibited PKA-mediated enhancement of RTA trans-activating capacity. Given the high catecholamine levels at sites of KSHV latency such as the vasculature and lymphoid organs, these data suggest that beta-adrenergic control of RTA might constitute a significant physiological regulator of KSHV lytic replication. These findings also suggest novel therapeutic strategies for controlling the activity of this oncogenic gammaherpesvirus in vivo.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transativadores/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Epinefrina/farmacologia , Proteínas Imediatamente Precoces/genética , Norepinefrina/farmacologia , Regiões Promotoras Genéticas , Transdução de Sinais , Transativadores/genética , Proteínas Virais/genética , Ativação Viral/efeitos dos fármacos
17.
Proc Natl Acad Sci U S A ; 101(23): 8727-32, 2004 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-15173593

RESUMO

Costimulation of purified CD8(+) T lymphocytes induces de novo expression of CD4, suggesting a previously unrecognized function for this molecule in the immune response. Here, we report that the CD4 molecule plays a direct role in CD8(+) T cell function by modulating expression of IFN-gamma and Fas ligand, two important CD8(+) T cell effector molecules. CD4 expression also allows infection of CD8 cells by HIV, which results in down-regulation of the CD4 molecule and impairs the induction of IFN-gamma, Fas ligand, and the cytotoxic responses of activated CD8(+) T cells. Thus, the CD4 molecule plays a direct role in CD8 T cell function, and infection of these cells by HIV provides an additional reservoir for the virus and also may contribute to the immunodeficiency seen in HIV disease.


Assuntos
Antígenos CD4/metabolismo , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/imunologia , Citotoxicidade Imunológica , Proteína Ligante Fas , Humanos , Técnicas In Vitro , Interferon gama/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Imunológicos , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA