Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Trends Cancer Res ; 13: 55-62, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30369774

RESUMO

Tumor suppressor function of Annexin-A7 (ANXA7) was demonstrated by cancer-prone phenotype in Anxa7(+/-) mice and ANXA7 profiling in human cancers including prostate and breast. Consistent with its more evident in vivo tumor suppressor role in prostate cancer, wild-type(wt)-ANXA7 in vitro induced similar G2-arrests, but reduced survival more drastically in prostate cancer cells compared to breast cancer cells (DU145 versus MDA-MB-231 and -435). In all three hormone-resistant cancer cell lines, wt-ANXA7 abolished the expression of the oncogenic low-molecular weight (LMW) cyclin E which was for the first time encountered in prostate cancer cells. Dominant-negative nMMM-ANXA7 (which lacks phosphatidylserine liposome aggregation properties) failed to abrogate LMW-cyclin E and simultaneously induced fibroblast growth factor 8 (FGF8) in DU145 that was consistent with the continuing cell cycle progression and reduced cell death. Adenoviral vector alone induced FGF8 in MDA-MB-231/435 cell lines, but not in DU145 cells. Our data indicated that the LMW-Cyclin E expressions in breast cancer and prostate cancer cell-lines were differentially regulated by wild-type and dominant-negative ANXA7 isoforms, demonstrating a different survival mechanism utilized by breast cancer cells. Conventional tumor suppressor p53 failed to completely abolish FGF8 and LMW-cyclin E in breast cancer cells, which were eventually translated into their survival. Thus, ANXA7 tumor suppression could modulate FGF8 and cyclin E expression, and control implying more specific associations with the annexin properties of ANXA7 in prostate tumorigenesis.

2.
Oncogene ; 29(17): 2457-66, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20190808

RESUMO

Annexin-A7 (ANXA7) tumor suppressor role has been shown in various tumors, and ANXA7 expression has been particularly lost in androgen-resistant prostate cancers. In this study, we studied ANXA7 regulation in normal prostate versus androgen-sensitive and -resistant prostate cancer cells. Deletion mapping analysis showed lowest ANXA7-promoter activities in androgen-sensitive LNCaP prostate cancer cells. Genomatix analysis of ANXA7 promoter identified a cluster of steroid nuclear hormone receptor elements, including V$GREF (V$GRE.02/ARE.02). Gelshift analysis clearly indicated distinct nuclear protein occupancy at this ANXA7-promoter site (-1086/-890) in prostate cancer (LNCaP, DU145, and PC3) versus normal prostate (PrEC) cells. In matrix-assisted laser desorption time-of-flight mass spectrometry-based search for ANXA7 nuclear regulators, we identified several heterogeneous nuclear ribonucleoproteins (hnRNPs) (A1, A2/B1 and K) attached to the steroid-associated ANXA7-promoter site in the androgen-resistant PC3 prostate cancer cells with high ANXA7 gene copy number, but not in PrEC. The hnPNP role in ANXA7 regulation (that was validated by hnRNPA2/B1 antibody interference) resulted in multiple ANXA7 cDNA and protein products in PC3, but not in PrEC. Ingenuity pathways analysis showed plausible molecular paths between ANXA7 and the hnRNP-associated network in prostate cancer progression. Thus, a multi-hnRNP complex can be responsible for aberrant ANXA7 transcription and splicing, thereby affecting ANXA7 expression pattern and tumor suppressor function in prostate cancer.


Assuntos
Anexina A7/genética , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Neoplasias da Próstata/genética , Proteínas Supressoras de Tumor/genética , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/fisiologia , Humanos , Masculino , Fosfatidilinositol 3-Quinases/fisiologia , Regiões Promotoras Genéticas , Próstata/metabolismo , Neoplasias da Próstata/prevenção & controle , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
3.
Biochim Biophys Acta ; 1742(1-3): 151-60, 2004 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-15590065

RESUMO

Mobilization of intracellular calcium from inositol-1,4,5-triphosphate (IP3)-sensitive endoplasmic reticulum (ER) stores plays a prominent role in brain function. Mice heterozygous for the annexin A7 (Anx7) gene have a profound reduction in IP3 receptor function in pancreatic islets along with defective insulin secretion. We examined IP3-sensitive calcium pools in the brains of Anx7 (+/-) mice by utilizing ATP/Mg(2+)-dependent (45)Ca(2+) uptake into brain membrane preparations and tissue sections. Although the Anx7 (+/-) mouse brain displayed similar levels of IP3 binding sites and thapsigargin-sensitive (45)Ca(2+) uptake as that seen in wild-type mouse brain, the Anx7 (+/-) mouse brain Ca(2+) pools showed markedly reduced sensitivity to IP3. A potent and saturable Ca(2+)-releasing effect of recombinant ANX7 protein was demonstrated in mouse and rat brain membrane preparations, which was additive with that of IP3. We propose that ANX7 mobilizes Ca(2+) from an endoplasmic reticulum-like pool, which can be recruited to enhance IP3-mediated Ca(2+) release.


Assuntos
Anexina A7/fisiologia , Encéfalo/metabolismo , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Animais , Anexina A7/genética , Anexina A7/metabolismo , Sequência de Bases , Primers do DNA , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley
4.
Dis Markers ; 17(2): 115-20, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11673658

RESUMO

The ANX7 gene codes for a Ca2+-activated GTPase, which has been implicated in both exocytotic secretion in cells and control of growth. In this review, we summarize information regarding increased tumor frequency in the Anx7 knockout mice, ANX7 growth suppression of human cancer cell lines, and ANX7 expression in human tumor tissue micro-arrays. The loss of ANX7 is significant in metastatic and hormone refractory prostate cancer compared to benign prostatic hyperplasia. In addition, ANX7 expression has prognostic value for predicting survival of breast cancer patients.


Assuntos
Anexina A7/metabolismo , Neoplasias da Mama/fisiopatologia , GTP Fosfo-Hidrolases/metabolismo , Neoplasias da Próstata/fisiopatologia , Animais , Anexina A7/genética , Biomarcadores Tumorais , Progressão da Doença , Feminino , GTP Fosfo-Hidrolases/genética , Humanos , Masculino
5.
Mol Med ; 7(8): 523-34, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11591888

RESUMO

BACKGROUND: Cystic fibrosis (CF) is the most common, lethal autosomal recessive disease affecting children in the United States and Europe. Extensive work is being performed to develop both gene and drug therapies. The principal mutation causing CF is in the CFTR gene ([Delta F508]CFTR). This mutation causes the mutant protein to traffic poorly to the plasma membrane, and degrades CFTR chloride channel activity. CPX, a candidate drug for CF, binds to mutant CFTR and corrects the trafficking deficit. CPX also activates mutant CFTR chloride channel activity. CF airways are phenotypically inundated by inflammatory signals, primarily contributed by sustained secretion of the proinflammatory cytokine interleukin 8 (IL-8) from mutant CFTR airway epithelial cells. IL-8 production is controlled by genes from the TNF-alphaR/NFkappaB pathway, and it is possible that the CF phenotype is due to dysfunction of genes from this pathway. In addition, because drug therapy with CPX and gene therapy with CFTR have the same common endpoint of raising the levels of CFTR, we have hypothesized that either approach should have a common genomic endpoint. MATERIALS AND METHODS: To test this hypothesis, we studied IL-8 secretion and global gene expression in IB-3 CF lung epithelial cells. The cells were treated by either gene therapy with wild-type CFTR, or by pharmacotherapy with the CFTR-surrogate drug CPX. CF cells, treated with either CFTR or CPX, were also exposed to Pseudomonas aeruginosa, a common chronic pathogen in CF patients. cDNA microarrays were used to assess global gene expression under the different conditions. A novel bioinformatic algorithm (GENESAVER) was developed to identify genes whose expression paralleled secretion of IL-8. RESULTS: We report here that IB3 CF cells secrete massive levels of IL-8. However, both gene therapy with CFTR and drug therapy with CPX substantially suppress IL-8 secretion. Nonetheless, both gene and drug therapy allow the CF cells to respond with physiologic secretion of IL-8 when the cells are exposed to P. aeruginosa. Thus, neither CFTR nor CPX acts as a nonspecific suppressor of IL-8 secretion from CF cells. Consistently, pharmacogenomic analysis indicates that CF cells treated with CPX greatly resemble CF cells treated with CFTR by gene therapy. Additionally, the same result obtains in the presence of P. aeruginosa. Classical hierarchical cluster analysis, based on similarity of global gene expression, also supports this conclusion. The GENESAVER algorithm, using the IL-8 secretion level as a physiologic variable, identifies a subset of genes from the TNF-alphaR/NFkappaB pathway that is expressed in phase with IL-8 secretion from CF epithelial cells. Certain other genes, previously known to be positively associated with CF, also fall into this category. Identified genes known to code for known inhibitors are expressed inversely, out of phase with IL-8 secretion. CONCLUSIONS: Wild-type CFTR and CPX both suppress proinflammatory IL-8 secretion from CF epithelial cells. The mechanism, as defined by pharmacogenomic analysis, involves identified genes from the TNF-alphaR/NFkappaB pathway. The close relationship between IL-8 secretion and genes from the TNF-alphaR/NFkappaB pathway suggests that molecular or pharmaceutical targeting of these novel genes may have strategic use in the development of new therapies for CF. From the perspective of global gene expression, both gene and drug therapy have similar genomic consequences. This is the first example showing equivalence of gene and drug therapy in CF, and suggests that a gene therapy-defined endpoint may prove to be a powerful paradigm for CF drug discovery. Finally, because the GENESAVER algorithm is capable of isolating disease-relevant genes in a hypothesis-driven manner without recourse to any a priori knowledge about the system, this new algorithm may also prove useful in applications to other genetic diseases.


Assuntos
Fibrose Cística/patologia , Interleucina-8/metabolismo , Pulmão/patologia , NF-kappa B/genética , Análise de Sequência com Séries de Oligonucleotídeos , Receptores do Fator de Necrose Tumoral/genética , Algoritmos , Linhagem Celular , Criança , Análise por Conglomerados , Fibrose Cística/genética , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação da Expressão Gênica , Terapia Genética , Humanos , Pulmão/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Pseudomonas aeruginosa/fisiologia , Receptores do Fator de Necrose Tumoral/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Fatores de Tempo , Xantinas/uso terapêutico
6.
Proc Natl Acad Sci U S A ; 98(8): 4575-80, 2001 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-11287641

RESUMO

The ANX7 gene is located on human chromosome 10q21, a site long hypothesized to harbor a tumor suppressor gene(s) (TSG) associated with prostate and other cancers. To test whether ANX7 might be a candidate TSG, we examined the ANX7-dependent suppression of human tumor cell growth, stage-specific ANX7 expression in 301 prostate specimens on a prostate tissue microarray, and loss of heterozygosity (LOH) of microsatellite markers at or near the ANX7 locus. Here we report that human tumor cell proliferation and colony formation are markedly reduced when the wild-type ANX7 gene is transfected into two prostate tumor cell lines, LNCaP and DU145. Consistently, analysis of ANX7 protein expression in human prostate tumor microarrays reveals a significantly higher rate of loss of ANX7 expression in metastatic and local recurrences of hormone refractory prostate cancer as compared with primary tumors (P = 0.0001). Using four microsatellite markers at or near the ANX7 locus, and laser capture microdissected tumor cells, 35% of the 20 primary prostate tumors show LOH. The microsatellite marker closest to the ANX7 locus showed the highest rate of LOH, including one homozygous deletion. We conclude that the ANX7 gene exhibits many biological and genetic properties expected of a TSG and may play a role in prostate cancer progression.


Assuntos
Anexina A7/genética , Genes Supressores de Tumor , Neoplasias da Próstata/genética , Divisão Celular/genética , Cromossomos Humanos Par 10 , Marcadores Genéticos , Humanos , Imuno-Histoquímica , Perda de Heterozigosidade , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Polimorfismo Genético , Neoplasias da Próstata/patologia
7.
Proc Natl Acad Sci U S A ; 96(24): 13783-8, 1999 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-10570150

RESUMO

The mammalian anx7 gene codes for a Ca(2+)-activated GTPase, which supports Ca(2+)/GTP-dependent secretion events and Ca(2+) channel activities in vitro and in vivo. To test whether anx7 might be involved in Ca(2+) signaling in secreting pancreatic beta cells, we knocked out the anx7 gene in the mouse and tested the insulin-secretory properties of the beta cells. The nullizygous anx7 (-/-) phenotype is lethal at embryonic day 10 because of cerebral hemorrhage. However, the heterozygous anx7 (+/-) mouse, although expressing only low levels of ANX7 protein, is viable and fertile. The anx7 (+/-) phenotype is associated with a substantial defect in insulin secretion, although the insulin content of the islets, is 8- to 10-fold higher in the mutants than in the normal littermate control. We infer from electrophysiological studies that both glucose-stimulated secretion and voltage-dependent Ca(2+) channel functions are normal. However, electrooptical recordings indicate that the (+/-) mutation has caused a change in the ability of inositol 1,4,5-trisphosphate (IP(3))-generating agonists to release intracellular calcium. The principle molecular consequence of lower anx7 expression is a profound reduction in IP(3) receptor expression and function in pancreatic islets. The profound increase in islets, beta cell number, and size may be a means of compensating for less efficient insulin secretion by individual defective pancreatic beta cells. This is a direct demonstration of a connection between glucose-activated insulin secretion and Ca(2+) signaling through IP(3)-sensitive Ca(2+) stores.


Assuntos
Anexina A7/fisiologia , Canais de Cálcio/biossíntese , Sinalização do Cálcio , GTP Fosfo-Hidrolases/fisiologia , Inositol 1,4,5-Trifosfato , Insulina/metabolismo , Receptores Citoplasmáticos e Nucleares/biossíntese , Animais , Anexina A7/genética , Cálcio/metabolismo , Linhagem Celular , Citosol , Eletrofisiologia , GTP Fosfo-Hidrolases/genética , Vetores Genéticos , Glucose/metabolismo , Hiperplasia , Hipertrofia , Imuno-Histoquímica , Receptores de Inositol 1,4,5-Trifosfato , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Knockout , Mutagênese , Fenótipo
8.
Cell Calcium ; 25(1): 59-68, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10191960

RESUMO

The role of intracellular calcium stores in stimulus-secretion coupling in the pancreatic beta-cell is largely unknown. We report here that tetracaine stimulates insulin secretion from collagenase-isolated mouse islets of Langerhans in the absence of glucose or extracellular calcium. We also found that the anesthetic evokes a dose-dependent rise of the intracellular free-calcium concentration ([Ca2+]i) in cultured rat and mouse beta-cells. The tetracaine-specific [Ca2+]i rise also occurs in the absence of glucose, or in beta-cells depolarized by exposure to a Ca(2+)-deficient medium (< 1 microM) or elevated [K+]o. Furthermore, tetracaine (> or = 300 microM) depolarized the beta-cell membrane in mouse pancreatic islets, but inhibited Ca2+ entry through voltage-gated Ca2+ channels in HIT cells, an insulin-secreting cell line. From these data we conclude that tetracaine-enhancement of insulin release occurs by mechanisms that are independent of Ca2+ entry across the cell membrane. The tetracaine-induced [Ca2+]i rise in cultured rat beta-cells and insulin secretion from mouse islets is insensitive to dantrolene (20 microM), a drug that inhibits Ca2+ release evoked by cholinergic agonists in the pancreatic beta-cell, and thapsigargin (3 microM), a blocker of the endoplasmic reticulum (ER) Ca2+ pump. We conclude that the Ca2+ required for tetracaine-potentiated insulin secretion is released from intracellular Ca2+ stores other than the ER. Furthermore, tetracaine-induced Ca2+ release was unaffected by the mitochondrial electron transfer inhibitors NaN3 and rotenone. Taken together, these data show that a calcium source other than the ER and mitochondria can affect beta-cell insulin secretion.


Assuntos
Cálcio/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Canais de Cálcio , Retículo Endoplasmático , Secreção de Insulina , Líquido Intracelular , Ativação do Canal Iônico , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Mitocôndrias , Ratos , Ratos Wistar , Tetracaína/metabolismo , Tetracaína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA