Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(10)2023 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-37240163

RESUMO

Annexin A7/ANXA7 is a calcium-dependent membrane fusion protein with tumor suppressor gene (TSG) properties, which is located on chromosome 10q21 and is thought to function in the regulation of calcium homeostasis and tumorigenesis. However, whether the molecular mechanisms for tumor suppression are also involved in the calcium- and phospholipid-binding properties of ANXA7 remain to be elucidated. We hypothesized that the 4 C-terminal endonexin-fold repeats in ANXA7 (GX(X)GT), which are contained within each of the 4 annexin repeats with 70 amino acids, are responsible for both calcium- and GTP-dependent membrane fusion and the tumor suppressor function. Here, we identified a dominant-negative triple mutant (DNTM/DN-ANXA7J) that dramatically suppressed the ability of ANXA7 to fuse with artificial membranes while also inhibiting tumor cell proliferation and sensitizing cells to cell death. We also found that the [DNTM]ANA7 mutation altered the membrane fusion rate and the ability to bind calcium and phospholipids. In addition, in prostate cancer cells, our data revealed that variations in phosphatidylserine exposure, membrane permeabilization, and cellular apoptosis were associated with differential IP3 receptor expression and PI3K/AKT/mTOR modulation. In conclusion, we discovered a triple mutant of ANXA7, associated with calcium and phospholipid binding, which leads to the loss of several essential functions of ANXA7 pertinent to tumor protection and highlights the importance of the calcium signaling and membrane fusion functions of ANXA7 for preventing tumorigenesis.


Assuntos
Fosfatidilinositol 3-Quinases , Neoplasias da Próstata , Masculino , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio , Neoplasias da Próstata/metabolismo , Proliferação de Células/genética , Linhagem Celular Tumoral , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Carcinogênese
2.
Ann N Y Acad Sci ; 971: 53-60, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12438089

RESUMO

The Anx7 gene codes for a Ca(2+)/GTPase with calcium channel and membrane fusion properties that has been proposed to regulate exocytotic secretion in chromaffin and other cell types. We have previously reported that the homozygous Anx7 (+/-) knockout mouse has an embryonically lethal phenotype. However, the viable heterozygous Anx7 (+/-) mouse displays a complex phenotype that includes adrenal gland hypertrophy, chromaffin cell hyperplasia, and defective IP(3) receptor (IP(3)R) expression. To search for a molecular basis for this phenotype, we have used cDNA microarray technology and have challenged control and mutant mice with fed or fasting conditions. We report that in the absence of the Anx7/IP(3)R signaling system, the cells in the adrenal gland are unable to discriminate between the fed and fasted states, in vivo. In control chromaffin cells, fasting is accompanied by an increased expression of structural genes for chromaffin cell contents, including chromogranin A and B, and DbetaH. There are also genes whose expression is specifically reduced. However, the Anx7 (+/-) mutation results in sustained expression of these nutritionally sensitive genes. We hypothesize that the calcium signaling defect due to the missing IP(3)R may be responsible for the global effects of the mutation on nutritionally sensitive genes. We further hypothesize that the tonically elevated expression of chromogranin A, a reportedly master control "switch" for dense core granule formation, may contribute to the process driving glandular hypertrophy and chromaffin cell hyperplasia in the Anx7 (+/-) mutant mouse.


Assuntos
Glândulas Suprarrenais/metabolismo , Anexina A7/genética , Anexina A7/fisiologia , Privação de Alimentos , Medula Suprarrenal/metabolismo , Fenômenos Fisiológicos da Nutrição Animal , Animais , Anexina A7/metabolismo , Canais de Cálcio/metabolismo , Células Cromafins/metabolismo , Cromogranina A , Cromograninas/metabolismo , DNA Complementar/metabolismo , Bases de Dados como Assunto , Hipertrofia , Receptores de Inositol 1,4,5-Trifosfato , Camundongos , Camundongos Knockout , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais
3.
Cancer Res ; 66(19): 9609-16, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17018618

RESUMO

Tumor suppressor function for Annexin A7 (ANXA7; 10q21) is based on cancer-prone phenotype in Anxa7(+/-) mouse and ANXA7 prognostic role in human cancers. Because ANXA7-caused liposome aggregation can be promoted by arachidonic acid (AA), we hypothesized that the phospholipid-binding tumor suppressor ANXA7 is associated with AA cascade. In a comparative study of ANXA7 versus canonical tumor suppressor p53 effects on AA lipoxygenation pathway in the p53-mutant and androgen-insensitive DU145 prostate cancer cells, both tumor suppressors altered gene expression of major 5-lipoxygenase (LOX) and 15-LOXs, including response to T helper 2 (Th2)-cytokine [interleukin-4 (IL-4)] and endogenous steroids (mimicked by dexamethasone). Wild-type and mutant ANXA7 distinctly affected expression of the dexamethasone-induced 15-LOX-2 (a prostate-specific endogenous tumor suppressor) as well as the IL-4-induced 15-LOX-1. On the other hand, wild-type p53 restored 5-LOX expression in DU145 to levels comparable to benign prostate epithelial cells. Using mass spectrometry of DNA affinity-enriched nuclear proteins, we detected different proteins that were bound to adjacent p53 and estrogen response elements in the 5-LOX promoter in DU145 cells introduced with ANXA7 versus p53. Sex hormone regulator 17-beta hydroxysteroid dehydrogenase 4 was identified under p53 introduction, which induced the 5-LOX expression. Meantime, nuclear proteins bound to the same 5-LOX promoter site under introduction of ANXA7 (that was associated with the repressed 5-LOX) were identified as zinc finger proteins ZNF433 and Aiolos, pyrin domain-containing NALP10, and the p53-regulating DNA repair enzyme APEX1. Thus, ANXA7 and p53 can distinctly regulate LOX transcription that is potentially relevant to the AA-mediated cell growth control in tumor suppression.


Assuntos
Adenocarcinoma/patologia , Anexina A7/fisiologia , Araquidonato 15-Lipoxigenase/biossíntese , Araquidonato 5-Lipoxigenase/biossíntese , Ácidos Araquidônicos/farmacologia , Proteínas de Neoplasias/fisiologia , Neoplasias da Próstata/patologia , Proteína Supressora de Tumor p53/fisiologia , Adenocarcinoma/metabolismo , Araquidonato 15-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/genética , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dexametasona/farmacologia , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes p53 , Humanos , Interleucina-4/farmacologia , Masculino , Próstata/citologia , Neoplasias da Próstata/metabolismo , Proteínas Recombinantes de Fusão/fisiologia
4.
Proc Natl Acad Sci U S A ; 102(27): 9631-6, 2005 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15983368

RESUMO

Digitoxin and structurally related cardiac glycoside drugs potently block activation of the TNF-alpha/NF-kappaB signaling pathway. We have hypothesized that the mechanism might be discovered by searching systematically for selective inhibitory action through the entire pathway. We report that the common action of these drugs is to block the TNF-alpha-dependent binding of TNF receptor 1 to TNF receptor-associated death domain. This drug action can be observed with native cells, such as HeLa, and reconstituted systems prepared in HEK293 cells. All other antiinflammatory effects of digitoxin on NF-kappaB and c-Jun N-terminal kinase pathways appear to follow from the blockade of this initial upstream signaling event.


Assuntos
Digitoxina/farmacologia , NF-kappa B/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/efeitos dos fármacos , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Western Blotting , Citometria de Fluxo , Células HeLa , Humanos , Imunoprecipitação , Luciferases , Plasmídeos/genética
5.
Mol Med ; 8(12): 781-97, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12606813

RESUMO

BACKGROUND: Gene expression in islets of Langerhans is profoundly sensitive to glucose and other nutrients. Islets of Langerhans in the Anx7(+/-) knockout mouse exhibit a profound reduction in ITPR3 protein expression, defective intracellular calcium signaling, and defective insulin secretion. Additional data presented here also show that mRNA for ITPR3 is virtually undetectable in isolated Anx7(+/-) islets. IP3Receptor type 3 (ITPR3) expression in islets of Langerhans is closely regulated by secretory stimuli, and it has been suggested that the level of the ITPR3 expression controls the ability of the islets to respond to nutritional signals. We report that although control islets respond to glucose in vitro by a transient increment in ITPR3 mRNA, the islets from the Anx7(+/-) mouse remain low. We therefore hypothesized that the Anx7/IP3 Receptor(3)/Ca(2+) signaling pathway plays a role in beta cell responses to glucose, and that in the absence of the Anx7/ITPR3 signaling system, the islets would be unable to discriminate between fed or fasted states in vivo. MATERIALS AND METHODS: To test this hypothesis, we subjected Anx7(+/-) and control mice to either food and water ad libidum or to an overnight fast with access to water only. We then isolated the respective islets and compared nutrient-dependent changes in global gene expression under the four conditions using genome-based microarray technology. RESULTS: Anx7 protein expression in these islets is only about 50% of control levels in normal littermate controls, and IPTR3 message and protein are virtually zero. cDNA microarray analyses show that in control animals gene expression is significantly affected by the fasting state. Many of the affected genes have historical relevance to development and differentiation of islets. These include preproglucagon, APOJ, cadherin2, phosphoglucoisomerase, oncostatin M, PAX6, HGF, and cytokeratin 18. However, there are also many other nutritionally sensitive genes in control islets that are principally associated with cell division and DNA repair. The latter genes have not specifically been associated with islet physiology in the past. By contrast, Anx7(+/-) mouse islets exhibit a greatly reduced ability to discriminate genomically between fed and fasted states for all classes of identified genes. Many of the validated genes are specific to islets in comparison to liver tissue examined. Real-time quantitative RT-PCR analysis of islets from Anx7 heterozygous mice and littermate controls revealed remarkable down-regulation in PTEN, Glut-2, PDX-1, IGF-1, and Neuro D1 expression, but not in liver. CONCLUSIONS: We conclude that reduced gene dosage in the Anx7(+/-) islet, with concomitant loss of ITPR3 expression and consequent defects in Ca(2+) signaling, may substantially contribute to the mechanism of the loss of genomic discrimination, in vivo, between the fed and fasted states. We believe that the requirement for complete Anx7 gene dosage and IPTR3 expression in islets of Langerhans will prove to be of fundamental importance for understanding the mechanism of nutritional sensing in health and disease.


Assuntos
Anexina A7/metabolismo , Regulação da Expressão Gênica/fisiologia , Ilhotas Pancreáticas/metabolismo , Transdução de Sinais/fisiologia , Algoritmos , Animais , Anexina A7/genética , Glicemia/metabolismo , Canais de Cálcio/biossíntese , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Receptores de Inositol 1,4,5-Trifosfato , Insulina/sangue , Insulina/metabolismo , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo
6.
Proc Natl Acad Sci U S A ; 100(24): 14287-92, 2003 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-14608035

RESUMO

Annexin 7 (ANX7) acts as a tumor suppressor gene in prostate cancer, where loss of heterozygosity and reduction of ANX7 protein expression is associated with aggressive metastatic tumors. To investigate the mechanism by which this gene controls tumor development, we have developed an Anx7(+/-) knockout mouse. As hypothesized, the Anx7(+/-) mouse has a cancer-prone phenotype. The emerging tumors express low levels of Anx7 protein. Nonetheless, the wild-type Anx7 allele is detectable in laser-capture microdissection-derived tumor tissue cells. Genome array analysis of hepatocellular carcinoma tissue indicates that the Anx7(+/-) genotype is accompanied by profound reductions of expression of several other tumor suppressor genes, DNA repair genes, and apoptosis-related genes. In situ analysis by tissue imprinting from chromosomes in the primary tumor and spectral karyotyping analysis of derived cell lines identify chromosomal instability and clonal chromosomal aberrations. Furthermore, whereas 23% of the mutant mice develop spontaneous neoplasms, all mice exhibit growth anomalies, including gender-specific gigantism and organomegaly. We conclude that haploinsufficiency of Anx7 expression appears to drive disease progression to cancer because of genomic instability through a discrete signaling pathway involving other tumor suppressor genes, DNA-repair genes, and apoptosis-related genes.


Assuntos
Anexina A7/genética , Genes Supressores de Tumor , Animais , Divisão Celular , Instabilidade Cromossômica , Dictyostelium/citologia , Dictyostelium/genética , Feminino , Dosagem de Genes , Expressão Gênica , Genes de Protozoários , Genótipo , Gigantismo/genética , Heterozigoto , Humanos , Cariotipagem , Neoplasias Hepáticas Experimentais/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Metástase Neoplásica/genética , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Neoplasias da Próstata/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA