Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunother ; 41(3): 130-140, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29239915

RESUMO

Chimeric antigen receptor (CAR) T cells represent a novel targeted approach to overcome both quantitative and qualitative shortfalls of the host immune system relating to the detection and subsequent destruction of tumors. The identification of antigens expressed specifically on the surface of tumor cells is a critical first step in the ability to utilize CAR T cells for the treatment of cancer. The 5T4 is a tumor-associated antigen which is expressed on the cell surface of most solid tumors including ovarian cancer. Matched blood and tumor samples were collected from 12 patients with ovarian cancer; all tumors were positive for 5T4 expression by immunohistochemistry. Patient T cells were effectively transduced with 2 different anti-5T4 CAR constructs which differed in their affinity for the target antigen. Co-culture of CAR T cells with matched autologous tumor disaggregates resulted in antigen-specific secretion of IFN-gamma. Furthermore, assessment of the efficacy of anti-5T4 CAR T cells in a mouse model resulted in therapeutic benefit against established ovarian tumors. These results demonstrate proof of principle that 5T4 is an attractive target for immune intervention in ovarian cancer and that patient T cells engineered to express a 5T4-specific CAR can recognize and respond physiologically to autologous tumor cells.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia Adotiva , Glicoproteínas de Membrana/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Imunoterapia Adotiva/métodos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Receptores de Antígenos Quiméricos/genética , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
2.
PLoS One ; 9(4): e94272, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24705452

RESUMO

Usher syndrome type 1B is a combined deaf-blindness condition caused by mutations in the MYO7A gene. Loss of functional myosin VIIa in the retinal pigment epithelia (RPE) and/or photoreceptors leads to blindness. We evaluated the impact of subretinally delivered UshStat, a recombinant EIAV-based lentiviral vector expressing human MYO7A, on photoreceptor function in the shaker1 mouse model for Usher type 1B that lacks a functional Myo7A gene. Subretinal injections of EIAV-CMV-GFP, EIAV-RK-GFP (photoreceptor specific), EIAV-CMV-MYO7A (UshStat) or EIAV-CMV-Null (control) vectors were performed in shaker1 mice. GFP and myosin VIIa expression was evaluated histologically. Photoreceptor function in EIAV-CMV-MYO7A treated eyes was determined by evaluating α-transducin translocation in photoreceptors in response to low light intensity levels, and protection from light induced photoreceptor degeneration was measured. The safety and tolerability of subretinally delivered UshStat was evaluated in macaques. Expression of GFP and myosin VIIa was confirmed in the RPE and photoreceptors in shaker1 mice following subretinal delivery of the EIAV-CMV-GFP/MYO7A vectors. The EIAV-CMV-MYO7A vector protected the shaker1 mouse photoreceptors from acute and chronic intensity light damage, indicated by a significant reduction in photoreceptor cell loss, and restoration of the α-transducin translocation threshold in the photoreceptors. Safety studies in the macaques demonstrated that subretinal delivery of UshStat is safe and well-tolerated. Subretinal delivery of EIAV-CMV-MYO7A (UshStat) rescues photoreceptor phenotypes in the shaker1 mouse. In addition, subretinally delivered UshStat is safe and well-tolerated in macaque safety studies These data support the clinical development of UshStat to treat Usher type 1B syndrome.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Vírus da Anemia Infecciosa Equina/genética , Síndromes de Usher/genética , Síndromes de Usher/terapia , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Ordem dos Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/metabolismo , Humanos , Macaca , Masculino , Camundongos , Camundongos Knockout , Miosina VIIa , Miosinas/genética , Fenótipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Transporte Proteico , Retina/metabolismo , Retina/patologia , Transducina/metabolismo
3.
J Cell Sci ; 123(Pt 1): 51-61, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20016066

RESUMO

The integrin family of heterodimeric cell-surface receptors are fundamental in cell-cell and cell-matrix adhesion. Changes to either integrin-ligand affinity or integrin gene expression are central to a variety of disease processes, including inflammation, cardiovascular disease and cancer. In screening for novel activators of integrin-ligand affinity we identified the previously uncharacterised multi-transmembrane domain protein Fam38A, located at the endoplasmic reticulum (ER). siRNA knockdown of Fam38A in epithelial cells inactivates endogenous beta1 integrin, reducing cell adhesion. Fam38A mediates integrin activation by recruiting the small GTPase R-Ras to the ER, which activates the calcium-activated protease calpain by increasing Ca(2+) release from cytoplasmic stores. Fam38A-induced integrin activation is blocked by inhibition of either R-Ras or calpain activity, or by siRNA knockdown of talin, a well-described calpain substrate. This highlights a novel mechanism for integrin activation by Fam38A, utilising calpain and R-Ras signalling from the ER. These data represent the first description of a novel spatial regulator of R-Ras, of an alternative integrin activation-suppression pathway based on direct relocalisation of R-Ras to the ER, and of a mechanism linking R-Ras and calpain signalling from the ER with modulation of integrin-ligand affinity.


Assuntos
Células Epiteliais/metabolismo , Integrina beta1/metabolismo , Proteínas de Membrana/metabolismo , Animais , Células CHO , Sinalização do Cálcio/genética , Calpaína/antagonistas & inibidores , Adesão Celular/genética , Cricetinae , Cricetulus , Retículo Endoplasmático , Células Epiteliais/citologia , Células HeLa , Humanos , Integrina beta1/genética , Canais Iônicos , Proteínas de Membrana/genética , Transporte Proteico/genética , RNA Interferente Pequeno/genética , Talina/genética , Proteínas ras/antagonistas & inibidores
4.
Mol Biol Cell ; 20(14): 3224-38, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19458194

RESUMO

The physical properties of the extracellular matrix (ECM) regulate the behavior of several cell types; yet, mechanisms by which cells recognize and respond to changes in these properties are not clear. For example, breast epithelial cells undergo ductal morphogenesis only when cultured in a compliant collagen matrix, but not when the tension of the matrix is increased by loading collagen gels or by increasing collagen density. We report that the actin-binding protein filamin A (FLNa) is necessary for cells to contract collagen gels, and pull on collagen fibrils, which leads to collagen remodeling and morphogenesis in compliant, low-density gels. In stiffer, high-density gels, cells are not able to contract and remodel the matrix, and morphogenesis does not occur. However, increased FLNa-beta1 integrin interactions rescue gel contraction and remodeling in high-density gels, resulting in branching morphogenesis. These results suggest morphogenesis can be "tuned" by the balance between cell-generated contractility and opposing matrix stiffness. Our findings support a role for FLNa-beta1 integrin as a mechanosensitive complex that bidirectionally senses the tension of the matrix and, in turn, regulates cellular contractility and response to this matrix tension.


Assuntos
Proteínas Contráteis/metabolismo , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Integrina beta1/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Fenômenos Biomecânicos , Linhagem Celular Tumoral , Colágeno/metabolismo , Filaminas , Géis/metabolismo , Humanos , Camundongos , Morfogênese , Cadeias Leves de Miosina/metabolismo , Fosforilação , Ligação Proteica
5.
Blood ; 112(13): 5130-40, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18799729

RESUMO

The ordered series of proliferation and differentiation from hematopoietic progenitor cells is disrupted in leukemia, resulting in arrest of differentiation at immature proliferative stages. Characterizing the molecular basis of hematopoietic differentiation is therefore important for understanding and treating disease. Retinoic acid induces expression of ankyrin repeat-containing protein with a suppressor of cytokine signaling box 2 (ASB2) in acute promyelocytic leukemia cells, and ASB2 expression inhibits growth and promotes commitment, recapitulating an early step critical for differentiation. ASB2 is the specificity subunit of an E3 ubiquitin ligase complex and is proposed to exert its effects by regulating the turnover of specific proteins; however, no ASB2 substrates had been identified. Here, we report that ASB2 targets the actin-binding proteins filamin A and B for proteasomal degradation. Knockdown of endogenous ASB2 in leukemia cells delays retinoic acid-induced differentiation and filamin degradation; conversely, ASB2 expression in leukemia cells induces filamin degradation. ASB2 expression inhibits cell spreading, and this effect is recapitulated by knocking down both filamin A and filamin B. Thus, we suggest that ASB2 may regulate hematopoietic cell differentiation by modulating cell spreading and actin remodeling through targeting of filamins for degradation.


Assuntos
Proteínas Contráteis/metabolismo , Leucemia/patologia , Proteínas dos Microfilamentos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Actinas/metabolismo , Adesão Celular , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas Contráteis/genética , Filaminas , Humanos , Leucemia/tratamento farmacológico , Proteínas dos Microfilamentos/genética , RNA Interferente Pequeno/farmacologia , Proteínas Supressoras da Sinalização de Citocina/genética , Tretinoína/farmacologia
6.
J Biol Chem ; 282(39): 28991-29001, 2007 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-17664272

RESUMO

Notch is a central regulator of important cell fate decisions. Notch activation produces diverse cellular effects suggesting the presence of context-dependent control mechanisms. Genetic studies have demonstrated that Notch and integrin mutations have related phenotypes in key developmental processes such as vascular development and somitogenesis. We show that the intracellular domain of mammalian Notch-1 activates integrins without affecting integrin expression. Integrin activation is dependent on gamma-secretase-mediated intramembranous cleavage of membrane-bound Notch releasing intracellular Notch that activates R-Ras, independent of CSL-transcription. Notch also reverses H-Ras and Raf-mediated integrin suppression without affecting ERK phosphorylation. Membrane-bound Notch mutants that are inefficiently cleaved or intracellular Notch mutants lacking the ankyrin repeat sequence do not activate R-Ras or integrins. Co-expression of Msx2-interacting nuclear target (MINT) protein with Notch or expression of intracellular Notch-1 truncation mutants lacking the C-terminal transactivation/PEST domain suppresses Notch transcriptional activity without affecting integrin activation. Notch ligand, Delta-like ligand-4, stimulates R-Ras-dependent alpha 5 beta 1 integrin-mediated adhesion, demonstrating the physiological relevance of this pathway. This new CSL-independent Notch/R-Ras pathway provides a molecular mechanism to explain Notch, integrin, and Ras cross-talk during the development of multicellular organisms.


Assuntos
Integrina alfa5beta1/metabolismo , Receptor Notch1/metabolismo , Proteínas ras/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Células CHO , Adesão Celular/fisiologia , Cricetinae , Cricetulus , Proteínas de Ligação a DNA , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Integrina alfa5beta1/genética , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas de Ligação a RNA , Receptor Notch1/genética , Proteínas ras/genética
7.
J Biol Chem ; 281(40): 29501-12, 2006 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-16895916

RESUMO

Phospholipase Cepsilon (PLCepsilon) is a newly described effector of the small GTP-binding protein H-Ras. Utilizing H-Ras effector mutants, we show that mutants H-Ras(G12V/E37G) and H-Ras(G12V/D38N) suppressed integrin activation in an ERK-independent manner. H-Ras(G12V/D38N) specifically activated the PLCepsilon effector pathway and suppressed integrin activation. Inhibition of PLCepsilon activation with a kinase-dead PLCepsilon mutant prevented H-Ras(G12V/D38N) from suppressing integrin activation, and low level expression of H-Ras(G12V/D38N) could synergize with wild-type PLCepsilon to suppress integrins. In addition, knockdown of endogenous PLCepsilon with small interfering RNA blocked H-Ras(G12V/D38N)-mediated integrin suppression. Suppressing integrin function with the H-Ras(G12V/D38N) mutant reduced cell adhesion to von Willebrand factor and fibronectin; this reduction in cell adhesion was blocked by coexpression of the kinase-dead PLCepsilon mutant. These results show that H-Ras suppresses integrin affinity via independent Raf and PLCepsilon signaling pathways and demonstrate a new physiological function for PLCepsilon in the regulation of integrin activation.


Assuntos
Regulação para Baixo , Integrinas/metabolismo , Fosfolipases Tipo C/fisiologia , Substituição de Aminoácidos/genética , Animais , Células CHO , Cricetinae , Cricetulus , Regulação para Baixo/genética , Glicina/genética , Fosfoinositídeo Fosfolipase C , Ratos , Serina/genética , Transdução de Sinais/genética , Treonina/genética , Fosfolipases Tipo C/genética , Valina/genética , Proteínas ras/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA