Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Mol Cancer ; 12: 90, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23919753

RESUMO

BACKGROUND: Recent epidemiological studies demonstrate that both active and involuntary exposure to tobacco smoke increase the risk of breast cancer. Little is known, however, about the molecular mechanisms by which continuous, long term exposure to tobacco smoke contributes to breast carcinogenesis because most previous studies have focused on short term treatment models. In this work we have set out to investigate the progressive transforming effects of tobacco smoke on non-tumorigenic mammary epithelial cells and breast cancer cells using in vitro and in vivo models of chronic cigarette smoke exposure. RESULTS: We show that both non-tumorigenic (MCF 10A, MCF-12A) and tumorigenic (MCF7) breast epithelial cells exposed to cigarette smoke acquire mesenchymal properties such as fibroblastoid morphology, increased anchorage-independent growth, and increased motility and invasiveness. Moreover, transplantation experiments in mice demonstrate that treatment with cigarette smoke extract renders MCF 10A cells more capable to survive and colonize the mammary ducts and MCF7 cells more prone to metastasize from a subcutaneous injection site, independent of cigarette smoke effects on the host and stromal environment. The extent of transformation and the resulting phenotype thus appear to be associated with the differentiation state of the cells at the time of exposure. Analysis by flow cytometry showed that treatment with CSE leads to the emergence of a CD44(hi)/CD24(low) population in MCF 10A cells and of CD44+ and CD49f + MCF7 cells, indicating that cigarette smoke causes the emergence of cell populations bearing markers of self-renewing stem-like cells. The phenotypical alterations induced by cigarette smoke are accompanied by numerous changes in gene expression that are associated with epithelial to mesenchymal transition and tumorigenesis. CONCLUSIONS: Our results indicate that exposure to cigarette smoke leads to a more aggressive and transformed phenotype in human mammary epithelial cells and that the differentiation state of the cell at the time of exposure may be an important determinant in the phenotype of the final transformed state.


Assuntos
Neoplasias da Mama/etiologia , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal , Fumar/efeitos adversos , Animais , Mama/metabolismo , Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Células-Tronco/metabolismo
2.
Biochem Biophys Res Commun ; 434(1): 70-4, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23545254

RESUMO

The high mobility group A1 gene (HMGA1) has been previously implicated in breast carcinogenesis, and is considered an attractive target for therapeutic intervention because its expression is virtually absent in normal adult tissue. Other studies have shown that knockdown of HMGA1 reduces the tumorigenic potential of breast cancer cells in vitro. Therefore, we sought to determine if silencing HMGA1 can affect breast cancer development and metastatic progression in vivo. We silenced HMGA1 expression in the human breast cancer cell line MDA-MB-231 using an RNA interference vector, and observed a significant reduction in anchorage-independent growth and tumorsphere formation, which respectively indicate loss of tumorigenesis and self-renewal ability. Moreover, silencing HMGA1 significantly impaired xenograft growth in immunodeficient mice, and while control cells metastasized extensively to the lungs and lymph nodes, HMGA1-silenced cells generated only a few small metastases. Thus, our results show that interfering with HMGA1 expression reduces the tumorigenic and metastatic potential of breast cancer cells in vivo, and lend further support to investigations into targeting HMGA1 as a potential treatment for breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Técnicas de Silenciamento de Genes , Proteína HMGA1a/antagonistas & inibidores , Proteína HMGA1a/genética , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Esferoides Celulares/patologia , Transplante Heterólogo/patologia
3.
Pancreatology ; 12(4): 372-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22898640

RESUMO

CONTEXT: Although pancreatic cancer is a common, highly lethal malignancy, the molecular events that enable precursor lesions to become invasive carcinoma remain unclear. We previously reported that the high-mobility group A1 (HMGA1) protein is overexpressed in >90% of primary pancreatic cancers, with absent or low levels in early precursor lesions. METHODS: Here, we investigate the role of HMGA1 in reprogramming pancreatic epithelium into invasive cancer cells. We assessed oncogenic properties induced by HMGA1 in non-transformed pancreatic epithelial cells expressing activated K-RAS. We also explored the HMGA1-cyclooxygenase (COX-2) pathway in human pancreatic cancer cells and the therapeutic effects of COX-2 inhibitors in xenograft tumorigenesis. RESULTS: HMGA1 cooperates with activated K-RAS to induce migration, invasion, and anchorage-independent cell growth in a cell line derived from normal human pancreatic epithelium. Moreover, HMGA1 and COX-2 expression are positively correlated in pancreatic cancer cell lines (r(2) = 0.93; p < 0.001). HMGA1 binds directly to the COX-2 promoter at an AT-rich region in vivo in three pancreatic cancer cell lines. In addition, HMGA1 induces COX-2 expression in pancreatic epithelial cells, while knock-down of HMGA1 results in repression of COX-2 in pancreatic cancer cells. Strikingly, we also discovered that Sulindac (a COX-1/COX-2 inhibitor) or Celecoxib (a more specific COX-2 inhibitor) block xenograft tumorigenesis from pancreatic cancer cells expressing high levels of HMGA1. CONCLUSIONS: Our studies identify for the first time an important role for the HMGA1-COX-2 pathway in pancreatic cancer and suggest that targeting this pathway could be effective to treat, or even prevent, pancreatic cancer.


Assuntos
Adenocarcinoma/genética , Ciclo-Oxigenase 2/genética , Proteína HMGA1a/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/prevenção & controle , Animais , Celecoxib , Divisão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Ciclo-Oxigenase 2/fisiologia , Inibidores de Ciclo-Oxigenase/administração & dosagem , Expressão Gênica , Proteína HMGA1a/fisiologia , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Transplante de Neoplasias , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/prevenção & controle , Pirazóis/administração & dosagem , Sulfonamidas/administração & dosagem , Sulindaco/administração & dosagem , Transplante Heterólogo , Proteínas ras/fisiologia
4.
BMC Genomics ; 12: 549, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-22053823

RESUMO

BACKGROUND: Although the high mobility group A1 (HMGA1) gene is widely overexpressed in diverse cancers and portends a poor prognosis in some tumors, the molecular mechanisms that mediate its role in transformation have remained elusive. HMGA1 functions as a potent oncogene in cultured cells and induces aggressive lymphoid tumors in transgenic mice. Because HMGA1 chromatin remodeling proteins regulate transcription, HMGA1 is thought to drive malignant transformation by modulating expression of specific genes. Genome-wide studies to define HMGA1 transcriptional networks during tumorigenesis, however, are lacking. To define the HMGA1 transcriptome, we analyzed gene expression profiles in lymphoid cells from HMGA1a transgenic mice at different stages in tumorigenesis. RESULTS: RNA from lymphoid samples at 2 months (before tumors develop) and 12 months (after tumors are well-established) was screened for differential expression of > 20,000 unique genes by microarray analysis (Affymetrix) using a parametric and nonparametric approach. Differential expression was confirmed by quantitative RT-PCR in a subset of genes. Differentially expressed genes were analyzed for cellular pathways and functions using Ingenuity Pathway Analysis. Early in tumorigenesis, HMGA1 induced inflammatory pathways with NFkappaB identified as a major node. In established tumors, HMGA1 induced pathways involved in cell cycle progression, cell-mediated immune response, and cancer. At both stages in tumorigenesis, HMGA1 induced pathways involved in cellular development, hematopoiesis, and hematologic development. Gene set enrichment analysis showed that stem cell and immature T cell genes are enriched in the established tumors. To determine if these results are relevant to human tumors, we knocked-down HMGA1 in human T-cell leukemia cells and identified a subset of genes dysregulated in both the transgenic and human lymphoid tumors. CONCLUSIONS: We found that HMGA1 induces inflammatory pathways early in lymphoid tumorigenesis and pathways involved in stem cells, cell cycle progression, and cancer in established tumors. HMGA1 also dyregulates genes and pathways involved in stem cells, cellular development and hematopoiesis at both early and late stages of tumorigenesis. These results provide insight into HMGA1 function during tumor development and point to cellular pathways that could serve as therapeutic targets in lymphoid and other human cancers with aberrant HMGA1 expression.


Assuntos
Transformação Celular Neoplásica/genética , Genes cdc , Proteína HMGA1a/metabolismo , Inflamação/genética , Tecido Linfoide/patologia , Células-Tronco/metabolismo , Transcriptoma , Animais , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/genética , Humanos , Leucemia de Células T/genética , Tecido Linfoide/metabolismo , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Neoplásico/genética
5.
Mod Pathol ; 23(1): 98-104, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19820691

RESUMO

Although pancreatic ductal adenocarcinoma is a common and almost uniformly fatal cancer, little is known about the molecular events that lead to tumor progression. The high-mobility group A1 (HMGA1) protein is an architectural transcription factor that has been implicated in the pathogenesis and progression of diverse human cancers, including pancreatic ductal adenocarcinoma. In this study, we investigated HMGA1 expression in pancreatic ductal adenocarcinoma cell lines and surgically resected tumors to determine whether it could be a marker for more advanced disease. By real-time quantitative RT-PCR, we measured HMGA1a mRNA in cultured pancreatic ductal adenocarcinoma cell lines and found increased levels in all cancer cells compared with normal pancreatic tissue. To investigate HMGA1 in primary human tumors, we performed immunohistochemical analysis of 125 cases of pancreatic adenocarcinoma and 99 precursor lesions (PanIN 1-3). We found nuclear staining for HMGA1 in 98% of cases of pancreatic adenocarcinoma, but only 43% of cases of PanIN precursor lesions. Moreover, HMGA1 immunoreactivity correlates positively with decreased survival and advanced tumor and PanIN grade. These results suggest that HMGA1 promotes tumor progression in pancreatic ductal adenocarcinoma and could be a useful biomarker and rational therapeutic target in advanced disease.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma Ductal Pancreático/patologia , Proteína HMGA1a/biossíntese , Neoplasias Pancreáticas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/mortalidade , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidade , Prognóstico , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos
6.
Mol Cancer Res ; 6(5): 743-50, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18505920

RESUMO

Although previous studies have established a prominent role for HMGA1 (formerly HMG-I/Y) in aggressive human cancers, the role of HMGA2 (formerly HMGI-C) in malignant transformation has not been clearly defined. The HMGA gene family includes HMGA1, which encodes the HMGA1a and HMGA1b protein isoforms, and HMGA2, which encodes HMGA2. These chromatin-binding proteins function in transcriptional regulation and recent studies also suggest a role in cellular senescence. HMGA1 proteins also appear to participate in cell cycle regulation and malignant transformation, whereas HMGA2 has been implicated primarily in the pathogenesis of benign, mesenchymal tumors. Here, we show that overexpression of HMGA2 leads to a transformed phenotype in cultured lung cells derived from normal tissue. Conversely, inhibiting HMGA2 expression blocks the transformed phenotype in metastatic human non-small cell lung cancer cells. Moreover, we show that HMGA2 mRNA and protein are overexpressed in primary human lung cancers compared with normal tissue or indolent tumors. In addition, there is a statistically significant correlation between HMGA2 protein staining by immunohistochemical analysis and tumor grade (P < 0.001). Our results indicate that HMGA2 is an oncogene important in the pathogenesis of human lung cancer. Although additional studies with animal models are needed, these findings suggest that targeting HMGA2 could be therapeutically beneficial in lung cancer and other cancers characterized by increased HMGA2 expression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína HMGA2/fisiologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Cromatina/metabolismo , Proteína HMGA2/metabolismo , Humanos , Imuno-Histoquímica/métodos , Fenótipo , RNA Mensageiro/metabolismo , Fatores de Tempo , Transcrição Gênica
7.
Cancer Res ; 67(9): 3998-4004, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17483309

RESUMO

Uterine cancer is the most common cancer of the female genital tract and is the fourth most frequent cause of cancer death in women in the U.S. Despite the high prevalence of uterine cancers, the molecular events that lead to neoplastic transformation in the uterus are poorly understood. Moreover, there are limited mouse models to study these malignancies. We generated transgenic mice with high-mobility group A1 gene (HMGA1a) expression targeted to uterine tissue and all female mice developed tumors by 9 months of age. Histopathologically, the tumors resemble human uterine adenosarcoma and are transplantable. To determine whether these findings are relevant to human disease, we evaluated primary human uterine neoplasms and found that HMGA1a mRNA and protein levels are increased in most high-grade neoplasms but not in normal uterine tissue, benign tumors, or most low-grade neoplasms. We also found that HMGA1a up-regulates cyclooxygenase 2 (COX-2) expression in transgenic tumors. Moreover, both HMGA1a and COX-2 expression are up-regulated in high-grade human leiomyosarcomas. Using chromatin immunoprecipitation, HMGA1a binds directly to the COX-2 promoter in human uterine cancer cells in vivo and activates its expression in transfection experiments. We also show that blocking either HMGA1a or COX-2 in high-grade human uterine cancer cells blocks anchorage-independent cell growth in methylcellulose. These findings show that HMGA1a functions as an oncogene when overexpressed in the uterus and contributes to the pathogenesis of human uterine cancer by activating COX-2 expression. Although a larger study is needed to confirm these results, HMGA1a may be a useful marker for aggressive human uterine cancers.


Assuntos
Adenossarcoma/genética , Ciclo-Oxigenase 2/biossíntese , Proteína HMGA1a/genética , Neoplasias Uterinas/genética , Adenossarcoma/enzimologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Ciclo-Oxigenase 2/genética , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/biossíntese , Humanos , Infertilidade Feminina/genética , Camundongos , Camundongos Nus , Camundongos Transgênicos , Transplante de Neoplasias , Transfecção , Regulação para Cima , Neoplasias Uterinas/enzimologia
8.
Mol Cancer Ther ; 7(7): 2090-5, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18645019

RESUMO

Uterine cancer is a common cause for cancer death in women and there is no effective therapy for metastatic disease. Thus, research is urgently needed to identify new therapeutic agents. We showed previously that all female HMGA1a transgenic mice develop malignant uterine tumors, indicating that HMGA1a causes uterine cancer in vivo. We also demonstrated that HMGA1a up-regulates cyclooxygenase-2 (COX-2) during tumorigenesis in this model. Similarly, we found that HMGA1a and COX-2 are overexpressed in human leiomyosarcomas, a highly malignant uterine cancer. Although epidemiologic studies indicate that individuals who take COX inhibitors have a lower incidence of some tumors, these inhibitors have not been evaluated in uterine cancer. Here, we show that HMGA1a mice on sulindac (a COX-1/COX-2 inhibitor) have significantly smaller uterine tumors than controls. To determine if COX inhibitors are active in human uterine cancers that overexpress HMGA1a, we treated cultured cells with sulindac sulfide or celecoxib (a specific COX-2 inhibitor). Both drugs block anchorage-independent growth in high-grade human uterine cancer cells that overexpress HMGA1a (MES-SA cells). In contrast, neither inhibitor blocked transformation in cells that do not overexpress HMGA1a. Moreover, xenograft tumors from MES-SA cells were significantly inhibited in mice on sulindac. More strikingly, no tumors formed in mice on celecoxib. These preclinical studies suggest that COX inhibitors could play a role in preventing tumor onset or progression in uterine cancers with dysregulation of the HMGA1a-COX-2 pathway. Importantly, these drugs have lower toxicity than chemotherapeutic agents used to treat advanced-stage uterine cancers.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Proteína HMGA1a/metabolismo , Neoplasias Uterinas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Celecoxib , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Nus , Camundongos Transgênicos , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Sulindaco/farmacologia
9.
Biochem Biophys Res Commun ; 365(1): 201-6, 2008 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17983591

RESUMO

Escherichia coli K1 is the most common Gram-negative organism causing neonatal meningitis. Binding to human brain microvascular endothelial cells (HBMEC) is an essential step for E. coli K1 traversal of the blood-brain barrier. In this study, we examined expression profiles of E. coli K1 strain RS218 during its binding to HBMEC. Comparison of HBMEC-bound E. coli K1 with collagen-bound E. coli revealed more than one hundred genes whose expression patterns were significantly changed in HBMEC-bound E. coli K1, but not in collagen-bound E. coli K1. These genes are involved mainly in cell surface decorations, cellular function, and nitrogen metabolism. The roles of several representative genes including frdA, clpB, carA, and ompT in HBMEC binding were verified with their isogenic mutants, which exhibited significantly less HBMEC binding capability compared to that of the parent strain. This transcriptome analysis provided us with the first genomic-level view of E. coli and HBMEC interactions.


Assuntos
Encéfalo/metabolismo , Endotélio Vascular/metabolismo , Escherichia coli/genética , Perfilação da Expressão Gênica , Escherichia coli/metabolismo , Humanos
10.
Eur Cytokine Netw ; 17(4): 260-5, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17353159

RESUMO

Microbial penetration of the blood-brain barrier (BBB) into the central nervous system is essential for the development of meningitis. Considerable progress has been achieved in understanding the pathophysiology of meningitis, however, relatively little is known about the early inflammatory events occurring at the time of bacterial crossing of the BBB. We investigated, using real-time quantitative PCR, the expression of the neutrophil chemoattractants alpha-chemokines CXCL1 (Groalpha) and CXCL8 (IL-8), and of the monocyte chemoattractant beta-chemokine CCL2 (MCP-1) by human brain microvascular endothelial cells (HBMEC) in response to the meningitis-causing E. coli K1 strain RS218 or its isogenic mutants lacking the ability to bind to and invade HBMEC. A nonpathogenic, laboratory E. coli strain HB101 was used as a negative control. CXCL8 was shown to be significantly expressed in HBMEC 4 hours after infection with E. coli K1, while no significant alterations were noted for CXCL1 and CCL2 expression. This upregulation of CXCL8 was induced by E. coli K1 strain RS218 and its derivatives lacking the ability to bind and invade HBMEC, but was not induced by the laboratory strain HB101. In contrast, no upregulation of CXCL8 was observed in human umbilical vein endothelial cells (HUVEC) after stimulation with E. coli RS218. These findings indicate that the CXCL8 expression is the result of the specific response of HBMEC to meningitis-causing E. coli K1.


Assuntos
Células Endoteliais/citologia , Escherichia coli/crescimento & desenvolvimento , Interleucina-8/genética , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Linhagem Celular , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CXCL1 , Quimiocinas CXC/genética , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Escherichia coli/classificação , Escherichia coli/genética , Regulação da Expressão Gênica , Humanos , Meningite devida a Escherichia coli/microbiologia , Mutação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Fatores de Tempo
11.
Res Microbiol ; 154(2): 105-14, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12648725

RESUMO

In this work we isolated a culturable endophytic aerobic heterotrophic bacterial community from the stem and root tissues of elm trees (Ulmus spp.) and analyzed its fluctuations. A total of 724 bacterial isolates were collected at different times (April, June, September and December) from two elm trees, one infected with Elm Yellows phytoplasmas, and one which was healthy-looking. The isolates were grouped into 82 haplotypes, identified by means of amplified ribosomal DNA restriction analysis (ARDRA) using the restriction enzyme AluI, suggesting that the genetic diversity of the bacterial community was very high. The taxonomic position of the isolates belonging to the twelve main haplotypes, representing more than 72% of the total population, was determined by 16S rDNA sequencing. The main genera were Bacillus, Curtobacterium, Pseudomonas, Stenotrophomonas, Sphingomonas, Enterobacter, and Staphylococcus. The fluctuations in the bacterial community, determined by different parameters (seasonal changes, plant organ, presence of phytoplasmas) were studied, revealing that they were influenced both by variations in temperature (warm or cold according to the season) and by the organ examined (roots or stems). The role of the phytopathogenic status in these fluctuations was also discussed.


Assuntos
Bactérias/isolamento & purificação , Ulmus/microbiologia , Bactérias/classificação , Bactérias/genética , Bactérias/patogenicidade , DNA Ribossômico/análise , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Ecossistema , Raízes de Plantas/microbiologia , Caules de Planta/microbiologia , RNA Ribossômico 16S/genética , Mapeamento por Restrição , Estações do Ano , Análise de Sequência de DNA
12.
Endothelium ; 11(5-6): 275-84, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15770770

RESUMO

Central nervous system (CNS) dysfunction is commonly observed in children with human immunodeficiency virus type 1 (HIV-1) infection, but the mechanism(s) whereby HIV-1 causes encephalopathy remains incompletely understood. Human brain microvascular endothelial cells (HBMECs), which constitute the blood-brain barrier, are likely to contribute to HIV-1 encephalopathy, but it is unclear whether HIV-1 receptors (CD4, chemokine receptors) are present on HBMECs. In the present study, the presence of CD4 in six different children was demonstrated. Moreover, the presence of CD4 in situ on brain sections was shown. Distribution of CD4 expression was heterogeneous among microvessels; staining for CD4 was strong in some vessels and absent in other adjacent vessels. CD4 and chemokine coreceptors were found to be functional as intercellular adhesion molecule (ICAM)-1 expression increased upon incubation of HBMECs with activating anti-CD4 and anti-chemokine receptor antibodies. The presence of CD4 and chemokine receptors in human brain endothelium of children may have implications for the pathogenesis of HIV-1 encephalopathy and explain the higher incidence of CNS involvement in HIV-1-infected children as compared to adults.


Assuntos
Encéfalo/metabolismo , Antígenos CD4/metabolismo , Células Endoteliais/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Células Endoteliais/virologia , Endotélio Vascular/metabolismo , Endotélio Vascular/virologia , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/patogenicidade , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
FEMS Microbiol Lett ; 230(1): 63-71, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14734167

RESUMO

A set of 146 Antarctic marine isolates from the Ross Sea was characterized by a combination of molecular techniques in order to determine the degree of inter- and intraspecific variability. Isolates were analyzed by amplified rDNA restriction analysis (ARDRA) using the tetrameric enzyme AluI, resulting in 52 different groups, corresponding to at least 52 different bacterial species, indicating a high degree of interspecific variability. The phylogenetic position of bacteria belonging to some ARDRA groups was obtained by sequencing of 16S rDNA. Random amplified polymorphic DNA (RAPD) analysis, carried out on the largest ARDRA groups, revealed a high intraspecific genetic variability, too. The analysis of plasmid content revealed the existence of horizontal gene transfer between strains belonging to the same and to different species. A comparison of the whole body of morphological, physiological and biochemical data was finally carried out.


Assuntos
Bactérias/classificação , Bactérias/genética , Biodiversidade , Temperatura Baixa , Água do Mar/microbiologia , Regiões Antárticas , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Meios de Cultura , DNA Bacteriano/análise , DNA Ribossômico/análise , Desoxirribonucleases de Sítio Específico do Tipo II , Dados de Sequência Molecular , Fenótipo , RNA Ribossômico 16S/genética , Técnica de Amplificação ao Acaso de DNA Polimórfico , Mapeamento por Restrição , Análise de Sequência de DNA
14.
PLoS One ; 8(7): e68630, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23844228

RESUMO

Downregulation of the tight junction protein claudin 1 is a frequent event in breast cancer and is associated with recurrence, metastasis, and reduced survival, suggesting a tumor suppressor role for this protein. Tumor suppressor genes are often epigenetically silenced in cancer. Downregulation of claudin 1 via DNA promoter methylation may thus be an important determinant in breast cancer development and progression. To investigate if silencing of claudin 1 has an epigenetic etiology in breast cancer we compared gene expression and methylation data from 217 breast cancer samples and 40 matched normal samples available through the Cancer Genome Atlas (TCGA). Moreover, we analyzed claudin 1 expression and methylation in 26 breast cancer cell lines. We found that methylation of the claudin 1 promoter CpG island is relatively frequent in estrogen receptor positive (ER+) breast cancer and is associated with low claudin 1 expression. In contrast, the claudin 1 promoter was not methylated in most of the ER-breast cancers samples and some of these tumors overexpress claudin 1. In addition, we observed that the demethylating agents, azacitidine and decitabine can upregulate claudin 1 expression in breast cancer cell lines that have a methylated claudin 1 promoter. Taken together, our results indicate that DNA promoter methylation is causally associated with downregulation of claudin 1 in a subgroup of breast cancer that includes mostly ER+ tumors, and suggest that epigenetic therapy to restore claudin 1 expression might represent a viable therapeutic strategy in this subtype of breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Claudina-1/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Linhagem Celular , Claudina-1/metabolismo , Análise por Conglomerados , Ilhas de CpG , Feminino , Ordem dos Genes , Inativação Gênica , Humanos , Transcriptoma
15.
Leuk Lymphoma ; 54(8): 1762-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23311322

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive leukemia with high relapse rates compared to B-lineage ALL. We previously showed that HMGA1a transgenic mice develop aggressive T-ALL, indicating that HMGA1 causes leukemic transformation in vivo. HMGA1 is also highly expressed in embryonic stem cells, hematopoietic stem cells and diverse, refractory human cancers. Disruption of the CDKN2A tumor suppressor locus occurs in most cases of T-ALL and is thought to contribute to leukemic transformation. To determine whether loss of function of CDKN2A cooperates with HMGA1 in T-ALL, we crossed HMGA1a transgenics onto a Cdkn2a null background. We discovered that T-ALL is markedly accelerated in HMGA1a transgenic Cdkn2a null mice. In addition, these mice recapitulate salient clinical and pathologic features of human T-ALL. HMGA1 is also highly overexpressed in human T-ALL. These findings suggest that HMGA1 plays a causative role in T-ALL and could represent a rational therapeutic target.


Assuntos
Transformação Celular Neoplásica/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Epistasia Genética , Inativação Gênica , Proteína HMGA1a/genética , Leucemia de Células T/genética , Animais , Modelos Animais de Doenças , Feminino , Expressão Gênica , Loci Gênicos , Humanos , Imunofenotipagem , Leucemia de Células T/metabolismo , Masculino , Camundongos , Camundongos Knockout
17.
Mol Cancer Res ; 7(11): 1803-12, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19903768

RESUMO

Although lung cancer is the leading cause of cancer death worldwide, the precise molecular mechanisms that give rise to lung cancer are incompletely understood. Here, we show that HMGA1 is an important oncogene that drives transformation in undifferentiated, large-cell carcinoma. First, we show that the HMGA1 gene is overexpressed in lung cancer cell lines and primary human lung tumors. Forced overexpression of HMGA1 induces a transformed phenotype with anchorage-independent cell growth in cultured lung cells derived from normal tissue. Conversely, inhibiting HMGA1 expression blocks anchorage-independent cell growth in the H1299 metastatic, undifferentiated, large-cell human lung carcinoma cells. We also show that the matrix metalloproteinase-2 (MMP-2) gene is a downstream target upregulated by HMGA1 in large-cell carcinoma cells. In chromatin immunoprecipitation experiments, HMGA1 binds directly to the MMP-2 promoter in vivo in large-cell lung cancer cells, but not in squamous cell carcinoma cells. In large-cell carcinoma cell lines, there is a significant, positive correlation between HMGA1 and MMP-2 mRNA. Moreover, interfering with MMP-2 expression blocks anchorage-independent cell growth in H1299 large-cell carcinoma cells, indicating that the HMGA1-MMP-2 pathway is required for this transformation phenotype in these cells. Blocking MMP-2 expression also inhibits migration and invasion in the H1299 large-cell carcinoma cells. Our findings suggest an important role for MMP-2 in transformation mediated by HMGA1 in large-cell, undifferentiated lung carcinoma and support the development of strategies to target this pathway in selected tumors.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Transformação Celular Neoplásica/metabolismo , Proteína HMGA1a/metabolismo , Neoplasias Pulmonares/metabolismo , Metaloproteinase 2 da Matriz/biossíntese , Carcinoma de Células Grandes/enzimologia , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/metabolismo , Carcinoma de Células Grandes/patologia , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/antagonistas & inibidores , Proteína HMGA1a/biossíntese , Proteína HMGA1a/genética , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transfecção , Regulação para Cima
19.
Cancer Res ; 68(24): 10121-7, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19074878

RESUMO

Although HMGA1 (high-mobility group A1; formerly HMG-I/Y) is an oncogene that is widely overexpressed in aggressive cancers, the molecular mechanisms underlying transformation by HMGA1 are only beginning to emerge. HMGA1 encodes the HMGA1a and HMGA1b protein isoforms, which function in regulating gene expression. To determine how HMGA1 leads to neoplastic transformation, we looked for genes regulated by HMGA1 using gene expression profile analysis. Here, we show that the STAT3 gene, which encodes the signaling molecule signal transducer and activator of transcription 3 (STAT3), is a critical downstream target of HMGA1a. STAT3 mRNA and protein are up-regulated in fibroblasts overexpressing HMGA1a and activated STAT3 recapitulates the transforming activity of HMGA1a in fibroblasts. HMGA1a also binds directly to a conserved region of the STAT3 promoter in vivo in human leukemia cells by chromatin immunoprecipitation and activates transcription of the STAT3 promoter in transfection experiments. To determine if this pathway contributes to HMGA1-mediated transformation, we investigated STAT3 expression in our HMGA1a transgenic mice, all of which developed aggressive lymphoid malignancy. STAT3 expression was increased in the leukemia cells from our transgenics but not in control cells. Blocking STAT3 function induced apoptosis in the transgenic leukemia cells but not in controls. In primary human leukemia samples, there was a positive correlation between HMGA1a and STAT3 mRNA. Moreover, blocking STAT3 function in human leukemia or lymphoma cells led to decreased cellular motility and foci formation. Our results show that the HMGA1a-STAT3 axis is a potential Achilles heel that could be exploited therapeutically in hematopoietic and other malignancies overexpressing HMGA1a.


Assuntos
Proteína HMGA1a/genética , Neoplasias Hematológicas/genética , Fator de Transcrição STAT3/genética , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Proteína HMGA1a/biossíntese , Proteína HMGA1a/metabolismo , Neoplasias Hematológicas/metabolismo , Humanos , Camundongos , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Fator de Transcrição STAT3/biossíntese , Fator de Transcrição STAT3/metabolismo , Transfecção , Regulação para Cima
20.
Alcohol Clin Exp Res ; 31(1): 130-7, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17207111

RESUMO

BACKGROUND: HIV-1 infection commonly leads to serious HIV-1-associated neurological disorders, such as HIV-1-associated encephalopathy and dementia. In addition, alcohol is commonly used and/or abused among AIDS patients, but it is unclear whether alcohol affects the disease progression and if it affects it, how this occurs. We hypothesized that alcohol could affect the blood-brain barrier (BBB) integrity and thus could affect the onset and/or progression of HIV-associated neurological disorders. METHODS: Human brain microvascular endothelial cells (HBMEC) in a BBB model system were pretreated with alcohol (17 and 68 mM) and subsequently coexposed with HIV-1 gp120. Expression of chemokine receptors CCR3, CCR5, and CXCR4 was assessed by enzyme-linked immunosorbent assay and real-time polymerase chain reaction. Changes in the permeability of the HBMEC monolayer were assessed using paracellular markers [(3)H]inulin or propidium iodide. Actin rearrangements in HBMEC were visualized by fluorescence microscopy and viability assessed using Live/Dead stain. RESULTS: Both gp120 and alcohol increased the permeability of the BBB model by up to 141%, without affecting HBMEC viability. Cotreatment with alcohol and gp120 did not result in a significant synergistic effect. Gp120 permeability involved chemokine receptor CCR5. Alcohol did not affect chemokine receptor expression on brain endothelial cells. Both gp120 and alcohol reorganized the cytoskeleton and induced stress fiber formation. Inhibition of reactive oxygen species (ROS) formation through NADPH blocked the effects of both gp120 and alcohol on permeability and stress fiber formation. CONCLUSION: These results indicate that both HIV-1 gp120 and alcohol induce stress fibers, causing increased permeability of the human BBB endothelium. Alcohol (68 mM)-mediated permeability increase was linked to ROS formation. The alcohol-mediated physiological changes in the HBMEC monolayers may increase diffusion of plasma components and viral penetration across the BBB. This suggests that alcohol, especially at levels attained in heavy drinkers, can potentially contribute in a negative fashion to HIV-1 neuropathogenesis.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Proteína gp120 do Envelope de HIV/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/patologia , Algoritmos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Indicadores e Reagentes , Receptores de Quimiocinas/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fibras de Estresse/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA