Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 315(4): G495-G510, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29848020

RESUMO

Lgr5-expressing intestinal stem cells (ISCs) maintain continuous and rapid generation of the intestinal epithelium. Here, we present evidence that dedifferentiation of committed enteroendocrine cells (EECs) contributes to maintenance of the epithelium under both basal conditions and in response to injury. Lineage-tracing studies identified a subset of EECs that reside at +4 position for more than 2 wk, most of which were BrdU-label-retaining cells. Under basal conditions, cells derived from these EECs grow from the bottom of the crypt to generate intestinal epithelium according to neutral drift kinetics that is consistent with dedifferentiation of mature EECs to ISCs. The lineage tracing of EECs demonstrated reserve stem cell properties in response to radiation-induced injury with the generation of reparative EEC-derived epithelial patches. Finally, the enterochromaffin (EC) cell was the predominant EEC type participating in these stem cell dynamics. These results provide novel insights into the +4 reserve ISC hypothesis, stem cell dynamics of the intestinal epithelium, and in the development of EC-derived small intestinal tumors. NEW & NOTEWORTHY The current manuscript demonstrating that a subset of mature enteroendocrine cells (EECs), predominantly enterochromaffin cells, dedifferentiates to fully functional intestinal stem cells (ISCs) is novel, timely, and important. These cells dedifferentiate to ISCs not only in response to injury but also under basal homeostatic conditions. These novel findings provide a mechanism in which a specified cell can dedifferentiate and contribute to normal tissue plasticity as well as the development of EEC-derived intestinal tumors under pathologic conditions.


Assuntos
Células-Tronco Adultas/citologia , Diferenciação Celular , Proliferação de Células , Células Enteroendócrinas/citologia , Intestino Delgado/citologia , Células-Tronco Adultas/metabolismo , Animais , Células Cultivadas , Células Enteroendócrinas/metabolismo , Intestino Delgado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Lesões Experimentais por Radiação/patologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
2.
Gastroenterology ; 153(6): 1555-1567.e15, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28859856

RESUMO

BACKGROUND & AIMS: The multiple endocrine neoplasia, type 1 (MEN1) locus encodes the nuclear protein and tumor suppressor menin. MEN1 mutations frequently cause neuroendocrine tumors such as gastrinomas, characterized by their predominant duodenal location and local metastasis at time of diagnosis. Diffuse gastrin cell hyperplasia precedes the appearance of MEN1 gastrinomas, which develop within submucosal Brunner's glands. We investigated how menin regulates expression of the gastrin gene and induces generation of submucosal gastrin-expressing cell hyperplasia. METHODS: Primary enteric glial cultures were generated from the VillinCre:Men1FL/FL:Sst-/- mice or C57BL/6 mice (controls), with or without inhibition of gastric acid by omeprazole. Primary enteric glial cells from C57BL/6 mice were incubated with gastrin and separated into nuclear and cytoplasmic fractions. Cells were incubated with forskolin and H89 to activate or inhibit protein kinase A (a family of enzymes whose activity depends on cellular levels of cyclic AMP). Gastrin was measured in blood, tissue, and cell cultures using an ELISA. Immunoprecipitation with menin or ubiquitin was used to demonstrate post-translational modification of menin. Primary glial cells were incubated with leptomycin b and MG132 to block nuclear export and proteasome activity, respectively. We obtained human duodenal, lymph node, and pancreatic gastrinoma samples, collected from patients who underwent surgery from 1996 through 2007 in the United States or the United Kingdom. RESULTS: Enteric glial cells that stained positive for glial fibrillary acidic protein (GFAP+) expressed gastrin de novo through a mechanism that required PKA. Gastrin-induced nuclear export of menin via cholecystokinin B receptor (CCKBR)-mediated activation of PKA. Once exported from the nucleus, menin was ubiquitinated and degraded by the proteasome. GFAP and other markers of enteric glial cells (eg, p75 and S100B), colocalized with gastrin in human duodenal gastrinomas. CONCLUSIONS: MEN1-associated gastrinomas, which develop in the submucosa, might arise from enteric glial cells through hormone-dependent PKA signaling. This pathway disrupts nuclear menin function, leading to hypergastrinemia and associated sequelae.


Assuntos
Duodeno/metabolismo , Gastrinas/metabolismo , Neuroglia/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Neoplasias Duodenais/enzimologia , Neoplasias Duodenais/genética , Neoplasias Duodenais/patologia , Duodeno/efeitos dos fármacos , Duodeno/patologia , Gastrinoma/enzimologia , Gastrinoma/genética , Gastrinoma/patologia , Gastrinas/genética , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Hiperplasia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroglia/efeitos dos fármacos , Inibidores de Proteassoma/farmacologia , Proteólise , Proteínas Proto-Oncogênicas/genética , Inibidores da Bomba de Prótons/farmacologia , Receptor de Colecistocinina B/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Fatores de Tempo , Ubiquitinação
3.
Gut ; 66(6): 1012-1021, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-26860771

RESUMO

BACKGROUND: Gastric carcinoids are slow growing neuroendocrine tumours arising from enterochromaffin-like (ECL) cells in the corpus of stomach. Although most of these tumours arise in the setting of gastric atrophy and hypergastrinemia, it is not understood what genetic background predisposes development of these ECL derived tumours. Moreover, diffuse microcarcinoids in the mucosa can lead to a field effect and limit successful endoscopic removal. OBJECTIVE: To define the genetic background that creates a permissive environment for gastric carcinoids using transgenic mouse lines. DESIGN: The multiple endocrine neoplasia 1 gene locus (Men1) was deleted using Cre recombinase expressed from the Villin promoter (Villin-Cre) and was placed on a somatostatin null genetic background. These transgenic mice received omeprazole-laced chow for 6 months. The direct effect of gastrin and the gastrin receptor antagonist YM022 on expression and phosphorylation of the cyclin inhibitor p27Kip1 was tested on the human human gastric adenocarcinoma cell line stably expressing CCKBR (AGSE) and mouse small intestinal neuroendocrine carcinoma (STC)-1 cell lines. RESULTS: The combination of conditional Men1 deletion in the absence of somatostatin led to the development of gastric carcinoids within 2 years. Suppression of acid secretion by omeprazole accelerated the timeline of carcinoid development to 6 months in the absence of significant parietal cell atrophy. Carcinoids were associated with hypergastrinemia, and correlated with increased Cckbr expression and nuclear export of p27Kip1 both in vivo and in gastrin-treated cell lines. Loss of p27Kip1 was also observed in human gastric carcinoids arising in the setting of atrophic gastritis. CONCLUSIONS: Gastric carcinoids require threshold levels of hypergastrinemia, which modulates p27Kip1 cellular location and stability.


Assuntos
Adenocarcinoma/metabolismo , Tumor Carcinoide/genética , Gastrinas/metabolismo , Proteínas Proto-Oncogênicas/genética , Somatostatina/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Adulto , Animais , Benzodiazepinas/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Feminino , Gastrinas/sangue , Gastrinas/genética , Gastrinas/farmacologia , Deleção de Genes , Antagonistas de Hormônios/farmacologia , Hormônios/farmacologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Omeprazol/farmacologia , Fosforilação/efeitos dos fármacos , Inibidores da Bomba de Prótons/farmacologia , RNA Mensageiro/metabolismo , Receptor de Colecistocinina B/genética , Receptor de Colecistocinina B/metabolismo , Transdução de Sinais
4.
Gastroenterology ; 144(7): 1466-77, 1477.e1-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23395646

RESUMO

BACKGROUND & AIMS: ZBP-89 (also ZNF148 or Zfp148) is a butyrate-inducible zinc finger transcription factor that binds to GC-rich DNA elements. Deletion of the N-terminal domain is sufficient to increase mucosal susceptibility to chemical injury and inflammation. We investigated whether conditional deletion of ZBP-89 from the intestinal and colonic epithelium of mice increases their susceptibility to pathogens such as Salmonella typhimurium. METHODS: We generated mice with a conditional null allele of Zfp148 (ZBP-89(FL/FL)) using homologous recombination to flank Zfp148 with LoxP sites (ZBP-89(FL/FL)), and then bred the resulting mice with those that express VillinCre. We used microarray analysis to compare gene expression patterns in colonic mucosa between ZBP-89(ΔInt) and C57BL/6 wild-type mice (controls). Mice were gavaged with 2 isogenic strains of S. typhimurium after administration of streptomycin. RESULTS: Microarray analysis revealed that the colonic mucosa of ZBP-89(ΔInt) mice had reduced levels of tryptophan hydroxylase 1 (Tph1) messenger RNA, encoding the rate-limiting enzyme in enterochromaffin cell serotonin (5-hydroxytryptamine [5HT]) biosynthesis. DNA affinity precipitation demonstrated direct binding of ZBP-89 to the mouse Tph1 promoter, which was required for its basal and butyrate-inducible expression. ZBP-89(ΔInt) mice did not increase mucosal levels of 5HT in response to S. typhimurium infection, and succumbed to the infection 2 days before control mice. The ΔhilA isogenic mutant of S. typhimurium lacks this butyrate-regulated locus and stimulated, rather than suppressed, expression of Tph1 approximately 50-fold in control, but not ZBP-89(ΔInt), mice, correlating with fecal levels of butyrate. CONCLUSIONS: ZBP-89 is required for butyrate-induced expression of the Tph1 gene and subsequent production of 5HT in response to bacterial infection in mice. Reductions in epithelial ZBP-89 increase susceptibility to colitis and sepsis after infection with S. typhimurium, partly because of reduced induction of 5HT production in response to butyrate and decreased secretion of antimicrobial peptides.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Mucosa Intestinal/imunologia , RNA Mensageiro/análise , Infecções por Salmonella/imunologia , Serotonina/biossíntese , Fatores de Transcrição/fisiologia , Triptofano Hidroxilase/fisiologia , Animais , Butiratos/imunologia , Colite/imunologia , Proteínas de Ligação a DNA/genética , Células Enterocromafins/imunologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Salmonella typhimurium , Serotonina/imunologia , Fatores de Transcrição/genética
5.
Am J Physiol Gastrointest Liver Physiol ; 303(6): G752-64, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22766853

RESUMO

Antral gastrin is the hormone known to stimulate acid secretion and proliferation of the gastric corpus epithelium. Patients with mutations in the multiple endocrine neoplasia type 1 (MEN1) locus, which encodes the protein menin, develop pituitary hyperplasia, insulinomas, and gastrinomas in the duodenum. We previously hypothesized that loss of menin leads to derepression of the gastrin gene and hypergastrinemia. Indeed, we show that menin represses JunD induction of gastrin in vitro. Therefore, we examined whether conditional deletion of Men1 (Villin-Cre and Lgr5-EGFP-IRES-CreERT2), with subsequent loss of menin from the gastrointestinal epithelium, increases gastrin expression. We found that epithelium-specific deletion of Men1 using Villin-Cre increased plasma gastrin, antral G cell numbers, and gastrin expression in the antrum, but not the duodenum. Moreover, the mice were hypochlorhydric by 12 mo of age, and gastric somatostatin mRNA levels were reduced. However, duodenal mRNA levels of the cyclin-dependent kinase inhibitor p27(Kip1) were decreased, and cell proliferation determined by Ki67 staining was increased. About 11% of the menin-deficient mice developed antral tumors that were negative for gastrin; however, gastrinomas were not observed, even at 12 mo of age. No gastrinomas were observed with conditional deletion of Men1 in the Lgr5 stem cells 5 mo after Cre induction. In summary, epithelium-specific deletion of the Men1 locus resulted in hypergastrinemia due to antral G cell hyperplasia and a hyperproliferative epithelium, but no gastrinomas. This result suggests that additional mutations in gene targets other than the Men1 locus are required to produce gastrin-secreting tumors.


Assuntos
Células Secretoras de Gastrina/fisiologia , Hiperplasia/patologia , Proteínas Proto-Oncogênicas/metabolismo , Antro Pilórico/citologia , Animais , Trato Gastrointestinal/metabolismo , Deleção de Genes , Regulação da Expressão Gênica/fisiologia , Genótipo , Hiperplasia/genética , Hiperplasia/metabolismo , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Somatostatina/metabolismo
6.
Oncotarget ; 8(55): 94330-94344, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29212231

RESUMO

ZBP-89 (Zfp148, ZNF148) is a Kruppel-type zinc-finger family transcription factor that binds to GC-rich DNA elements. Earlier studies in cell lines demonstrated that ZBP-89 cooperates with Wnt ß-catenin signaling by inducing ß-catenin gene expression. Since ß-catenin levels are normally highest at the crypt base, we examined whether ZBP-89 is required for stem cell maintenance. Lineage-tracing using a Zfp148CreERT2 transgenic line demonstrated expression in both intestine and colonic stem cells. Deleting the Zfp148 locus in the colon using the Cdx2NLSCreERT2 transgene, reduced the size and number of polyps formed in the Apc-deleted mice. Since colon polyps form in the presence of butyrate, a short chain fatty acid that suppresses cell growth, we examined the direct effect of butyrate on colon organoid survival. Butyrate induced senescence of colon organoids carrying the Apc deletion, only when Zfp148 was deleted. Using quantitative PCR and chromatin immunoprecipitation, we determined that butyrate treatment of colon cell lines suppressed ZNF148 gene expression, inducing CDKN2a (p16Ink4a ) gene expression. Collectively, Zfp148 mRNA is expressed in CBCs, and is required for stem cell maintenance and colonic transformation. Butyrate induces colonic cell senescence in part through suppression of ZBP-89 gene expression and its subsequent occupancy of the CDKN2A promoter.

7.
J Clin Invest ; 126(8): 2867-80, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27427984

RESUMO

Chronic Helicobacter pylori infection triggers neoplastic transformation of the gastric mucosa in a small subset of patients, but the risk factors that induce progression to gastric metaplasia have not been identified. Prior to cancer development, the oxyntic gastric glands atrophy and are replaced by metaplastic cells in response to chronic gastritis. Previously, we identified schlafen 4 (Slfn4) as a GLI1 target gene and myeloid differentiation factor that correlates with spasmolytic polypeptide-expressing metaplasia (SPEM) in mice. Here, we tested the hypothesis that migration of SLFN4-expressing cells from the bone marrow to peripheral organs predicts preneoplastic changes in the gastric microenvironment. Lineage tracing in Helicobacter-infected Slfn4 reporter mice revealed that SLFN4+ cells migrated to the stomach, where they exhibited myeloid-derived suppressor cell (MDSC) markers and acquired the ability to inhibit T cell proliferation. SLFN4+ MDSCs were not observed in infected GLI1-deficient mice. Overexpression of sonic hedgehog ligand (SHH) in infected WT mice accelerated the appearance of SLFN4+ MDSCs in the gastric corpus. Similarly, in the stomachs of H. pylori-infected patients, the human SLFN4 ortholog SLFN12L colocalized to cells that expressed MDSC surface markers CD15+CD33+HLA-DRlo. Together, these results indicate that SLFN4 marks a GLI1-dependent population of MDSCs that predict a shift in the gastric mucosa to a metaplastic phenotype.


Assuntos
Proteínas de Transporte/metabolismo , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Células Supressoras Mieloides/metabolismo , Animais , Sítios de Ligação , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Proteínas de Transporte/genética , Diferenciação Celular , Movimento Celular , Proteínas Hedgehog/metabolismo , Helicobacter pylori , Humanos , Interferon Tipo I/metabolismo , Ligantes , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Estômago/patologia , Linfócitos T/citologia , Tioglicolatos/química
8.
Cancer Res ; 76(23): 6877-6887, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27758879

RESUMO

In colorectal cancer, APC-mediated induction of unregulated cell growth involves posttranslational mechanisms that prevent proteasomal degradation of proto-oncogene ß-catenin (CTNNB1) and its eventual translocation to the nucleus. However, about 10% of colorectal tumors also exhibit increased CTNNB1 mRNA. Here, we show in colorectal cancer that increased expression of ZNF148, the gene coding for transcription factor ZBP-89, correlated with reduced patient survival. Tissue arrays showed that ZBP-89 protein was overexpressed in the early stages of colorectal cancer. Conditional deletion of Zfp148 in a mouse model of Apc-mediated intestinal polyps demonstrated that ZBP-89 was required for polyp formation due to induction of Ctnnb1 gene expression. Chromatin immunoprecipitation (ChIP) and EMSA identified a ZBP-89-binding site in the proximal promoter of CTNNB1 Reciprocally, siRNA-mediated reduction of CTNNB1 expression also decreased ZBP-89 protein. ChIP identified TCF DNA binding sites in the ZNF148 promoter through which Wnt signaling regulates ZNF148 gene expression. Suppression of either ZNF148 or CTNNB1 reduced colony formation in WNT-dependent, but not WNT-independent cell lines. Therefore, the increase in intracellular ß-catenin protein initiated by APC mutations is sustained by ZBP-89-mediated feedforward induction of CTNNB1 mRNA. Cancer Res; 76(23); 6877-87. ©2016 AACR.


Assuntos
Neoplasias Colorretais/genética , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética , beta Catenina/metabolismo , Animais , Neoplasias Colorretais/patologia , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Proto-Oncogene Mas , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transfecção
9.
FEMS Immunol Med Microbiol ; 44(3): 277-82, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15907449

RESUMO

We examined Mycoplasma penetrans-specific antibodies in sera of five male homosexual AIDS patients from whom M. penetrans was isolated during the disease process. No consistent immune reaction pattern could be recognized in Western blot using whole cell proteins. Serum samples obtained prior to M. penetrans isolation reacted with a number of M. penetrans proteins, most likely due to non-specific cross-reactions. Further analysis revealed that patients produced prominent antibody reaction to lipid-associated membrane proteins (LAMPs) of M. penetrans at the time of mycoplasma isolation, which could not be observed for serum samples obtained prior to M. penetrans isolation. The positive antibody reaction was mainly directed against two major LAMPs of M. penetrans with molecular mass of 35 and 38 kDa and produced a distinctive pattern of positive immunoreaction bands. Our observation suggested that, comparing with whole mycoplasmal proteins, LAMPs were more specific target antigens in serological assays for M. penetrans infection.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/sangue , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Infecções por Mycoplasma/sangue , Mycoplasma penetrans/imunologia , Especificidade de Anticorpos , Antígenos de Bactérias/análise , Proteínas da Membrana Bacteriana Externa/análise , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Masculino , Mycoplasma penetrans/isolamento & purificação
10.
Clin Infect Dis ; 36(10): 1246-53, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12746769

RESUMO

Lipid-associated membrane proteins (LAMPs) from 14 Mycoplasma hominis isolates or strains share similar protein and antigenicity profiles. Of 31 human immunodeficiency virus-infected patients from whose samples M. hominis was cultured, 28 tested strongly positive for serum antibodies to M. hominis LAMPs. The remaining 3 serum samples showed low antibody titer to LAMPs from all of the 14 M. hominis isolates or strains, which was likely the result of the compromised immune systems of the patients. Thus, M. hominis LAMPs as a whole are homogenous in antigenicity within the species, despite having many different serotypes. Serological study involving 564 healthy blood donors and 211 patients attending sexually transmitted disease clinics by LAMPs showed that general populations were widely exposed to M. hominis. Women were infected with M. hominis at a younger age than were men. The prevalence of infection increased markedly among sexually active persons.


Assuntos
Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Infecções por Mycoplasma/diagnóstico , Mycoplasma hominis/isolamento & purificação , Anticorpos Antibacterianos/classificação , Especificidade de Anticorpos , Antígenos de Bactérias/análise , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Metabolismo dos Lipídeos , Masculino , Proteínas de Membrana/análise , Infecções por Mycoplasma/sangue , Infecções por Mycoplasma/imunologia , Infecções por Mycoplasma/microbiologia , Estudos Soroepidemiológicos
11.
PLoS One ; 8(3): e58935, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23520544

RESUMO

Chronic inflammation in the stomach induces metaplasia, the pre-cancerous lesion that precedes inflammation-driven neoplastic transformation. While Hedgehog signaling contributes to the initiation of some cancers, its role in gastric transformation remains poorly defined. We found that Helicobacter-infected C57BL/6 mice develop extensive mucous cell metaplasia at 6 month but not at 2 months post-infection. Gastric metaplasia coincided with the appearance of CD45(+)MHCII(+)CD11b(+)CD11c(+) myeloid cells that were normally not present in the chronic gastritis at 2 months. The myeloid regulatory gene Schlafen-4 was identified in a microarray analysis comparing infected WT versus Gli1 null mice and was expressed in the CD11b(+)CD11c(+) myeloid population. Moreover this same population expressed IL-1ß and TNFα pro-inflammatory cytokines. By 6 months, the mucous neck cell metaplasia (SPEM) expressed IL-6, phosphorylated STAT3 and the proliferative marker Ki67. Expression was not observed in Gli1 mutant mice consistent with the requirement of Gli1 to induce this pre-neoplastic phenotype. Ectopic Shh ligand expression alone was not sufficient to induce SPEM, but with Helicobacter infection synergistically increased the histologic severity observed with the inflammation. Therefore Hedgehog signaling is required, but is not sufficient to generate pre-neoplastic changes during chronic gastritis. Gli1-dependent myeloid cell differentiation plays a pivotal role in the appearance of myeloid cell subtypes ostensibly required for SPEM development. Moreover, it suggests that therapies capable of targeting this phenotypic switch might prevent progression to metaplasia, the pre-neoplastic change that develops prior to dysplasia and gastric cancer, which also occurs in other epithelial-derived neoplasias initiated by chronic inflammation.


Assuntos
Gastrite/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter felis , Fatores de Transcrição Kruppel-Like/metabolismo , Células Mieloides/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Doença Crônica , Citocinas/biossíntese , Citocinas/genética , Gastrite/genética , Gastrite/microbiologia , Gastrite/patologia , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Infecções por Helicobacter/genética , Infecções por Helicobacter/patologia , Infecções por Helicobacter/prevenção & controle , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Metaplasia , Camundongos , Camundongos Transgênicos , Células Mieloides/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Proteína GLI1 em Dedos de Zinco
12.
J Biol Chem ; 277(19): 16913-9, 2002 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-11847231

RESUMO

Previous studies have demonstrated that cyclopentenone prostaglandins (cyPG) inhibit human immunodeficiency virus type 1 (HIV-1) replication in various cell types. We investigated the role of PG in the replication of HIV-1 in primary macrophages. The cyPG, PGA(1) and PGA(2), inhibited HIV-1 replication in acutely infected human monocyte-derived macrophages (MDM). Because PGA(1) and PGA(2) have previously been shown to be peroxisome proliferator-activated receptor gamma (PPARgamma) agonists, we examined the effect of synthetic PPARgamma agonists on HIV replication. The PPARgamma agonist ciglitazone inhibited HIV-1 replication in a dose-dependent manner in acutely infected human MDM. In addition, cyPG and ciglitazone reduced HIV replication in latently infected and viral entry-independent U1 cells, suggesting an effect at the level of HIV gene expression. Ciglitazone also suppressed HIV-1 mRNA levels as measured by reverse transcriptase PCR, in parallel with the decrease in reverse transcriptase activity. Co-transfection of PPARgamma wild type vectors and treatment with PPARgamma agonists inhibited HIV-1 promoter activity in U937 cells. Activation of PPARgamma also decreased HIV-1 mRNA stability following actinomycin D treatment. In summary, our experimental findings implicate PPARgamma as an important factor in the suppression of HIV-1 gene expression in MDM by cyPG. Thus natural and synthetic PPARgamma agonists may play a role in controlling HIV-1 infection in macrophages.


Assuntos
HIV-1/metabolismo , Macrófagos/metabolismo , Processamento Pós-Transcricional do RNA , Receptores Citoplasmáticos e Nucleares/agonistas , Tiazolidinedionas , Fatores de Transcrição/agonistas , Transcrição Gênica , Linhagem Celular , Células Cultivadas , Dactinomicina/farmacologia , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Hipoglicemiantes/farmacologia , Macrófagos/virologia , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Prostaglandinas A/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , DNA Polimerase Dirigida por RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiazóis/farmacologia , Fatores de Tempo , Transfecção , Células U937
13.
Cell Immunol ; 215(1): 61-71, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12142037

RESUMO

Since macrophages are a source of increased PGE(2) in AIDS, we investigated the role of PGE(2) in the replication of HIV-1 in these cells. PGE(2) inhibited HIV-1 replication measured by reverse transcriptase in human monocyte-derived macrophage (MDM). Treatment of MDM with the PGE(1) analog misoprostol, the adenylate cyclase activator forskolin, and the cyclic AMP analog dibutyryl-cyclic AMP (db-cAMP) suppressed HIV replication. The protein kinase A (PKA) activator 8-bromo-cyclic AMP also inhibited HIV-1 replication. Similar results were observed with the entry-independent, latently HIV-infected U1 cells. There was a parallel decrease in HIV-1 mRNA levels following PGE(2) treatment. Co-transfection of the HIV-1 promoter LTR.luciferase, with the vector CMV.Calpha, which expresses the PKA catalytic unit increasing PKA activity, reduced HIV-1 promoter activity. Inhibition of PKA activity with the pMT.RAB vector, a mutant regulatory unit of PKA, augmented HIV-1 promoter activity. In summary, PGE(2) inhibits HIV-1 gene expression in MDM through a PKA-dependent mechanism.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprostona/farmacologia , HIV-1/efeitos dos fármacos , Macrófagos/virologia , Replicação Viral/efeitos dos fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Inibidores de Adenilil Ciclases , Bucladesina/farmacologia , Células Cultivadas , Colforsina/farmacologia , Dinoprostona/biossíntese , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , HIV-1/genética , HIV-1/metabolismo , Humanos , Cinética , Macrófagos/enzimologia , Macrófagos/imunologia , Misoprostol/farmacologia , Monócitos/fisiologia , Regiões Promotoras Genéticas , RNA Viral/biossíntese , Receptores CCR5/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa