Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Cell ; 149(4): 847-59, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22541070

RESUMO

Alu RNA accumulation due to DICER1 deficiency in the retinal pigmented epithelium (RPE) is implicated in geographic atrophy (GA), an advanced form of age-related macular degeneration that causes blindness in millions of individuals. The mechanism of Alu RNA-induced cytotoxicity is unknown. Here we show that DICER1 deficit or Alu RNA exposure activates the NLRP3 inflammasome and triggers TLR-independent MyD88 signaling via IL18 in the RPE. Genetic or pharmacological inhibition of inflammasome components (NLRP3, Pycard, Caspase-1), MyD88, or IL18 prevents RPE degeneration induced by DICER1 loss or Alu RNA exposure. These findings, coupled with our observation that human GA RPE contains elevated amounts of NLRP3, PYCARD, and IL18 and evidence of increased Caspase-1 and MyD88 activation, provide a rationale for targeting this pathway in GA. Our findings also reveal a function of the inflammasome outside the immune system and an immunomodulatory action of mobile elements.


Assuntos
Elementos Alu , RNA Helicases DEAD-box/metabolismo , Atrofia Geográfica/imunologia , Atrofia Geográfica/patologia , Inflamassomos/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Ribonuclease III/metabolismo , Animais , Proteínas de Transporte/metabolismo , Atrofia Geográfica/metabolismo , Humanos , Inflamassomos/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Epitélio Pigmentado da Retina/patologia , Receptores Toll-Like/metabolismo
2.
Cell Commun Signal ; 18(1): 15, 2020 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-31987042

RESUMO

BACKGROUND: We have previously shown that the zinc finger transcription repressor SNAI2 (SLUG) represses tumor suppressor BRCA2-expression in non-dividing cells by binding to the E2-box upstream of the transcription start site. However, it is unclear how proliferating breast cancer (BC) cells that has higher oxidation state, overcome this repression. In this study, we provide insight into the mechanism of de-silencing of BRCA2 gene expression by PRDX5A, which is the longest member of the peroxiredoxin5 family, in proliferating breast cancer cells. METHODS: We used cell synchronization and DNA affinity pulldown to analyze PRDX5A binding to the BRCA2 silencer. We used oxidative stress and microRNA (miRNA) treatments to study nuclear localization of PRDX5A and its impact on BRCA2-expression. We validated our findings using mutational, reporter assay, and immunofluorescence analyses. RESULTS: Under oxidative stress, proliferating BC cells express PRDX5 isoform A (PRDX5A). In the nucleus, PRDX5A binds to the BRCA2 silencer near the E2-box, displacing SLUG and enhancing BRCA2-expression. Nuclear PRDX5A is translated from the second AUG codon in frame to the first AUG codon in the PRDX5A transcript that retains all exons. Mutation of the first AUG increases nuclear localization of PRDX5A in MDA-MB-231 cells, but mutation of the second AUG decreases it. Increased mitronic hsa-miRNA-6855-3p levels under oxidative stress renders translation from the second AUG preferable. Mutational analysis using reporter assay uncovered a miR-6855-3p binding site between the first and second AUG codon in the PRDX5A transcript. miR-6855-3p mimic increases accumulation of nuclear PRDX5A and inhibits reporter gene translation. CONCLUSION: Oxidative stress increases miR-6855-3p expression and binding to the inter-AUG sequence of the PRDX5A transcript, promoting translation of nuclear PRDX5A. Nuclear PRDX5A relieves SLUG-mediated BRCA2 silencing, resulting in increased BRCA2-expression.


Assuntos
Proteína BRCA2/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Inativação Gênica , MicroRNAs/metabolismo , Peroxirredoxinas/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Proteína BRCA2/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Feminino , Humanos , MicroRNAs/genética , Oxirredução , Estresse Oxidativo , Peroxirredoxinas/genética , Ligação Proteica , Transporte Proteico , Fatores de Transcrição da Família Snail/genética
3.
Circ Res ; 119(5): 621-34, 2016 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-27354210

RESUMO

RATIONALE: Gamma aminobutyric acid (GABA), a neurotransmitter of the central nervous system, is found in the systemic circulation of humans at a concentration between 0.5 and 3 µmol/L. However, the potential source of circulating GABA and its significance on the vascular system remains unknown. We hypothesized that endothelial cells (ECs) may synthesize and release GABA to modulate some functions in the EC and after its release into the circulation. OBJECTIVE: To assess whether GABA is synthesized and released by the EC and its potential functions. METHODS AND RESULTS: Utilizing the human umbilical vein ECs and aortic ECs, we demonstrated for the first time that ECs synthesize and release GABA from [1-(14)C]glutamate. Localization of GABA and the presence of the GABA-synthesizing enzyme, glutamic acid decarboxylase in EC were confirmed by immunostaining and immunoblot analysis, respectively. The presence of GABA was further confirmed by immunohistochemistry in the EC lining the human coronary vessel. EC-derived GABA regulated the key mechanisms of ATP synthesis, fatty acid, and pyruvate oxidation in EC. GABA protected EC by inhibiting the reactive oxygen species generation and prevented monocyte adhesion by attenuating vascular cell adhesion molecule -1 and monocyte chemoattractant protein-1 expressions. GABA had no relaxing effect on rat aortic rings. GABA exhibited a dose-dependent fall in blood pressure. However, the fall in BP was abolished after pretreatment with pentolinium. CONCLUSIONS: Our findings indicate novel potential functions of endothelium-derived GABA.


Assuntos
Células Endoteliais/metabolismo , Ácido gama-Aminobutírico/biossíntese , Ácido gama-Aminobutírico/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Ácido gama-Aminobutírico/farmacologia
4.
Nature ; 471(7338): 325-30, 2011 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-21297615

RESUMO

Geographic atrophy (GA), an untreatable advanced form of age-related macular degeneration, results from retinal pigmented epithelium (RPE) cell degeneration. Here we show that the microRNA (miRNA)-processing enzyme DICER1 is reduced in the RPE of humans with GA, and that conditional ablation of Dicer1, but not seven other miRNA-processing enzymes, induces RPE degeneration in mice. DICER1 knockdown induces accumulation of Alu RNA in human RPE cells and Alu-like B1 and B2 RNAs in mouse RPE. Alu RNA is increased in the RPE of humans with GA, and this pathogenic RNA induces human RPE cytotoxicity and RPE degeneration in mice. Antisense oligonucleotides targeting Alu/B1/B2 RNAs prevent DICER1 depletion-induced RPE degeneration despite global miRNA downregulation. DICER1 degrades Alu RNA, and this digested Alu RNA cannot induce RPE degeneration in mice. These findings reveal a miRNA-independent cell survival function for DICER1 involving retrotransposon transcript degradation, show that Alu RNA can directly cause human pathology, and identify new targets for a major cause of blindness.


Assuntos
Elementos Alu/genética , RNA Helicases DEAD-box/deficiência , Degeneração Macular/genética , Degeneração Macular/patologia , RNA/genética , RNA/metabolismo , Ribonuclease III/deficiência , Animais , Morte Celular , Sobrevivência Celular , Células Cultivadas , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Camundongos , MicroRNAs/metabolismo , Dados de Sequência Molecular , Oligonucleotídeos Antissenso , Fenótipo , Epitélio Pigmentado da Retina/enzimologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Ribonuclease III/genética , Ribonuclease III/metabolismo
5.
Arch Biochem Biophys ; 604: 95-102, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27311614

RESUMO

In this study, we have identified cystathionine (CTH), a sulfur containing metabolite, to be selectively enriched in human breast cancer (HBC) tissues (∼50-100 pmoles/mg protein) compared with undetectable levels in normal breast tissues. The accumulation of CTH, specifically in HBC, was attributed to the overexpression of cystathionine beta synthase (CBS), its synthesizing enzyme, and the undetectable levels of its downstream metabolizing enzyme, cystathionine gamma lyase (CGL). Interestingly both CBS and CGL could not be detected in normal breast tissues. We further observed that CTH protected HBC cells against excess reactive oxygen species (ROS) and chemotherapeutic drug-induced apoptosis. Moreover, CTH promoted both mitochondrial and endoplasmic reticulum homeostasis in HBC cells. As both the mitochondria and the endoplasmic reticulum are key organelles regulating the onset of apoptosis, we reasoned that endogenous CTH could be contributing towards increasing the apoptotic threshold in HBC cells. An increased apoptotic threshold is a hallmark of all cancer types, including HBC, and is primarily responsible for drug resistance. Hence this study unravels one of the possible pathways that may contribute towards drug resistance in HBC.


Assuntos
Neoplasias da Mama/metabolismo , Cistationina beta-Sintase/metabolismo , Cistationina gama-Liase/metabolismo , Cistationina/metabolismo , Resistencia a Medicamentos Antineoplásicos , Antineoplásicos/química , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Células MCF-7 , Microscopia Eletrônica , Consumo de Oxigênio , Permeabilidade , Espécies Reativas de Oxigênio/metabolismo
6.
BMC Cancer ; 16: 559, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27473585

RESUMO

BACKGROUND: Breast cancer is a complex heterogeneous disease where many distinct subtypes are found. Younger African American (AA) women often present themselves with aggressive form of breast cancer with unique biology which is very difficult to treat. Better understanding the biology of AA breast tumors could lead to development of effective treatment strategies. Our previous studies indicate that AA but not Caucasian (CA) triple negative (TN) breast cancer cells were sensitive to nitrosative stress-induced cell death. In this study, we elucidate possible mechanisms that contribute to nitric oxide (NO)-induced apoptosis in AA TN breast cancer cells. METHODS: Breast cancer cells were treated with various concentrations of long-acting NO donor, DETA-NONOate and cell viability was determined by trypan blue exclusion assay. Apoptosis was determined by TUNEL and caspase 3 activity as well as changes in mitochondrial membrane potential. Caspase 3 and Bax cleavage, levels of Cu/Zn superoxide dismutase (SOD) and Mn SOD was assessed by immunoblot analysis. Inhibition of Bax cleavage by Calpain inhibitor, and levels of reactive oxygen species (ROS) as well as SOD activity was measured in NO-induced apoptosis. In vitro and in vivo effect of NO treatment on mammary cancer stem cells (MCSCs) was assessed. RESULTS AND DISCUSSION: NO induced mitocondria-mediated apoptosis in all AA but not in CA TN breast cancer cells. We found significant TUNEL-positive cells, cleavage of Bax and caspase-3 activation as well as depolarization mitochondrial membrane potential only in AA TN breast cancer cells exposed to NO. Inhibition of Bax cleavage and quenching of ROS partially inhibited NO-induced apoptosis in AA TN cells. Increase in ROS coincided with reduction in SOD activity in AA TN breast cancer cells. Furthermore, NO treatment of AA TN breast cancer cells dramatically reduced aldehyde dehydrogenase1 (ALDH1) expressing MCSCs and xenograft formation but not in breast cancer cells from CA origin. CONCLUSIONS: Ethnic differences in breast tumors dictate a need for tailoring treatment options more suited to the unique biology of the disease.


Assuntos
Apoptose/efeitos dos fármacos , Compostos Nitrosos/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto , Negro ou Afro-Americano , Família Aldeído Desidrogenase 1 , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Immunoblotting , Isoenzimas/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Nus , Mitocôndrias/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Retinal Desidrogenase/metabolismo , Superóxido Dismutase/metabolismo , Neoplasias de Mama Triplo Negativas/etnologia , Neoplasias de Mama Triplo Negativas/metabolismo , Proteína X Associada a bcl-2/metabolismo
7.
Adv Exp Med Biol ; 814: 69-75, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25015801

RESUMO

Gestational diabetes, pre-eclampsia as well as intra-uterine infection during pregnancy affects the function of the endothelium both in the mother and the fetus leading to endothelial dysfunction. Gestational diabetes is also associated with an increased incidence of pre-eclampsia and it is likely that both the hyperglycemia as well as the release of cytokines especially TNFα during hyperglycemia may play an important role in the pathogenesis of endothelial dysfunction leading to preeclampsia. Similarly, some but not all studies have suggested that infection of the mother under certain circumstances can also lead to preeclampsia as women with either a bacterial or viral infection were at a higher risk of developing preeclampsia, compared to women without infection and infection also leads to a release in TNFα. Endothelial cells exposed to either high glucose or TNFα leads to an increase in the production of H2O2 and to a decrease in endothelial cell proliferation. The cellular and molecular mechanisms involved in this phenomenon are discussed.Gestational diabetes, pre-eclampsia as well as intra-uterine infection during pregnancy has profound effects on the fetus and long term effects on the neonate. All three conditions affect the function of the endothelium both in the mother and the fetus leading to endothelial dysfunction. Gestational diabetes is also associated with an increased incidence of pre-eclampsia and it is likely that both the hyperglycemia as well as the release of cytokines especially TNFα during hyperglycemia may play an important role in the pathogenesis of endothelial dysfunction leading to preeclampsia. It has also been suggested although not universally accepted that under certain circumstances maternal infection may also predispose to pre-eclampsia. Pre-eclampsia is also associated with the release of TNFα and endothelial dysfunction. However, the cellular and molecular mechanism(s) leading to the endothelial dysfunction by either hyperglycemia or by the cytokine TNFα appear to be different. In this chapter, we explore some of the similarities and differences leading to endothelial dysfunction by both hyperglycemia and by the inflammatory cytokine TNFα and the cellular and molecular mechanism(s) involved.


Assuntos
Citocinas/imunologia , Diabetes Gestacional/imunologia , Endotélio Vascular/imunologia , Pré-Eclâmpsia/imunologia , Complicações Infecciosas na Gravidez/imunologia , Diabetes Gestacional/metabolismo , Endotélio Vascular/metabolismo , Feminino , Humanos , Pré-Eclâmpsia/metabolismo , Gravidez , Complicações Infecciosas na Gravidez/metabolismo
8.
J Biol Chem ; 287(23): 19472-86, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22496452

RESUMO

One of highly pathogenic breast cancer cell types are the triple negative (negative in the expression of estrogen, progesterone, and ERBB2 receptors) breast cancer cells. These cells are highly motile and metastatic and are characterized by high levels of the metastasis regulator protein SLUG. Using isogenic breast cancer cell systems we have shown here that high motility of these cells is directly correlated with the levels of the SLUG in these cells. Because epithelial/mesenchymal cell motility is known to be negatively regulated by the catenin protein plakoglobin, we postulated that the transcriptional repressor protein SLUG increases the motility of the aggressive breast cancer cells through the knockdown of the transcription of the plakoglobin gene. We found that SLUG inhibits the expression of plakoglobin gene directly in these cells. Overexpression of SLUG in the SLUG-deficient cancer cells significantly decreased the levels of mRNA and protein of plakoglobin. On the contrary, knockdown of SLUG in SLUG-high cancer cells elevated the levels of plakoglobin. Blocking of SLUG function with a double-stranded DNA decoy that competes with the E2-box binding of SLUG also increased the levels of plakoglobin mRNA, protein, and promoter activity in the SLUG-high triple negative breast cancer cells. Overexpression of SLUG in the SLUG-deficient cells elevated the motility of these cells. Knockdown of plakoglobin in these low motility non-invasive breast cancer cells rearranged the actin filaments and increased the motility of these cells. Forced expression of plakoglobin in SLUG-high cells had the reverse effects on cellular motility. This study thus implicates SLUG-induced repression of plakoglobin as a motility determinant in highly disseminating breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Movimento Celular , Desmoplaquinas/biossíntese , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Desmoplaquinas/genética , Feminino , Humanos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , gama Catenina
9.
Arch Biochem Biophys ; 540(1-2): 33-40, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24157690

RESUMO

It is established that high concentrations of nitric oxide(1) (NO), as released from activated macrophages, induce apoptosis in breast cancer cells. In this study, we assessed the potential of a light-activated NO donor [(Me2bpb)Ru(NO)(Resf)], a recently reported apoptototic agent, in suppressing the anchorage independent growth potentials of an aggressive human breast cancer cell line. Our results demonstrated the down regulation of anchorage independent growth by light activated NO treatment in the aggressive human breast cancer cell line MDA-MB-231 and afforded insight into the associated mechanism(s). The investigation revealed an up-regulation of the bioactivity of catalase with an accompanied reduction in the endogenous levels of H2O2, a direct substrate of catalase and a recently identified endogenous growth modulator in breast cancer cells. An earlier publication reported that endogenous superoxide (O2(-)) in human breast cancer cells constitutively inhibits catalase bioactivity (at the level of its protein), resulting in increased H2O2 levels. Interestingly in this study, O2(-) was also found to be down- regulated following NO treatment providing a basis for the observed increase in catalase bioactivity. Cells silenced for the catalase gene exhibited compromised reduction in anchorage independent growth upon light activated NO treatment. Collectively this study detailed a mechanistic cross talk between exogenous NO and endogenous ROS in attenuating anchorage independent growth.


Assuntos
Peróxido de Hidrogênio/metabolismo , Luz , Óxido Nítrico/metabolismo , Compostos Organometálicos/farmacologia , Superóxidos/metabolismo , Neoplasias da Mama/patologia , Catalase/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias , Humanos , Óxido Nítrico/química , Compostos Organometálicos/química , Compostos Organometálicos/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Superóxido Dismutase/metabolismo , Tirosina/análogos & derivados , Tirosina/biossíntese
10.
J Biol Chem ; 286(1): 469-79, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21044962

RESUMO

UbcH5c, a member of the UbcH5 family of protein ubiquitin conjugase E2 enzymes, is a critical component of biological processes in human cells, being the initial ubiquitinating enzyme of substrates like IκB, TP53, and cyclin D1. We report here that the metastasis regulator protein SLUG inhibits the expression of UbcH5c directly through chromatin remodeling and thus, among other downstream effects, elevates the level of cyclin D1, thus enhancing the growth rates of breast cancer cells. Overexpression of SLUG in the SLUG-deficient breast cancer cells significantly decreased the levels of mRNA and protein of UbcH5c but only elevated the protein levels of cyclin D1. On the contrary, knockdown of SLUG in SLUG-high breast cancer cells elevated the levels of UbcH5c while decreasing the level of cyclin D1 protein. SLUG is recruited at the E2-box sequence at the UbcH5c gene promoter along with the corepressor CtBP1 and the effector HDAC1 to silence the expression of this gene. Knockdown of UbcH5c in the SLUG-deficient human breast cells elevated the level of cyclin D1 as well as the rates of proliferation and invasiveness of these cells. Whereas the growth rates of the cells are enhanced due to overexpression of SLUG or knockdown of UbcH5c in the breast cancer cells tested, ER(+) cells also acquire resistance to the anti-estrogen 4-hydroxytamoxifen due to the rise of cyclin D1 levels in these cells. This study thus implicates high levels of SLUG and low levels of UbcH5c as a determinant in the progression of metastatic breast cancer.


Assuntos
Neoplasias da Mama/patologia , Ciclina D1/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitinação , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Montagem e Desmontagem da Cromatina , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Invasividade Neoplásica/genética , Regiões Promotoras Genéticas/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Fatores de Transcrição da Família Snail , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Enzimas de Conjugação de Ubiquitina/deficiência , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação/genética
11.
J Biol Chem ; 286(8): 6614-26, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21149457

RESUMO

The parasitic protozoan Leishmania invades mammalian macrophages to establish infection. We reported previously that Leishmania manipulates the expression of several non-coding RNA genes (e.g. Alu RNA, B1 RNA, and signal recognition particle RNA) in macrophages to favor the establishment of their infection in the phagolysosomes of these cells (Ueda, Y., and Chaudhuri, G. (2000) J. Biol. Chem. 275, 19428-19432; Misra, S., Tripathi, M. K., and Chaudhuri, G. (2005) J. Biol. Chem. 280, 29364-29373). We report here the mechanism of this down-regulation. We found that the non-coding RNA (ncRNA) genes that are repressed by Leishmania infection in macrophages contain a "B-box" in their promoters and thus require the polymerase III transcription factor TFIIIC for their expression. We also found that Leishmania promastigotes through their surface protease (leishmanolysin or gp63) activate the thrombin receptor PAR1 in the macrophages. This activation of PAR1 raised the cytosolic concentration of Ca(2+) into the micromolar range, thereby activating the Ca(2+)-dependent protease µ-calpain. µ-Calpain then degraded TFIIIC110 to inhibit the expression of the selected ncRNA genes. Avirulent stocks of Leishmania not expressing surface gp63 failed to down-regulate ncRNAs in the exposed macrophages. Inhibition of PAR1 or calpain 1 in macrophages made them resistant to Leishmania infection. These data suggest that macrophage PAR1 and calpain 1 are potential drug targets against leishmaniasis.


Assuntos
Regulação para Baixo , Leishmania major/metabolismo , Leishmaniose Cutânea/metabolismo , Macrófagos/metabolismo , RNA Polimerase III/metabolismo , RNA não Traduzido/biossíntese , Animais , Cálcio/metabolismo , Calpaína/genética , Calpaína/metabolismo , Linhagem Celular Tumoral , Humanos , Leishmania major/genética , Leishmania major/patogenicidade , Leishmaniose Cutânea/genética , Macrófagos/parasitologia , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , RNA Polimerase III/genética , RNA não Traduzido/genética , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Fatores de Transcrição TFIII/genética , Fatores de Transcrição TFIII/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 31(11): 2586-94, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21903942

RESUMO

OBJECTIVE: Tumor necrosis factor-α (TNFα) may change from a stimulator of reversible activation of endothelial cells (ECs) to a killer when combined with cycloheximide (CHX). The means by which endothelial cells are destined to either the survival pathway or the apoptotic pathway are not fully understood. We investigated the role of p38 mitogen-activated protein kinase (MAPK) and protein phosphatase 2A (PP2A) activation and their regulation of 4E-BP1 stability in ECs to determine whether this pathway contributes to apoptosis induced by TNFα and CHX. METHODS AND RESULTS: Apoptosis was induced in human umbilical vein ECs (HUVECs) by treating them with a combination of TNFα and CHX (TNFα/CHX). Activation of p38 MAPK was increased in HUVECs undergoing apoptosis, which was associated with degradation of eukaryotic initiation factor 4A regulator 4E-BP1 in a p38 MAPK-dependent manner. CHX attenuated a TNFα-stimulated increase in the expression and activity of PP2A. Silencing PP2A expression with small interfering RNA transfection mimicked CHX sensitization, increasing HUVEC apoptosis with TNFα stimulation and suggesting a protective role for PP2A in the apoptotic process. CONCLUSION: Our data suggest that (1) TNFα stimulates PP2A and HUVECs elude apoptosis by PP2A-dependent dephosphorylation of p38 MAPK, and (2) CHX-induced inhibition of PP2A leads to maintenance of p38 activity and degradation of 4E-BP1, resulting in enhanced TNFα-induced apoptosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fosfoproteínas/metabolismo , Proteína Fosfatase 2/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteínas de Ciclo Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Cicloeximida/farmacologia , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fator de Iniciação 4E em Eucariotos/metabolismo , Humanos , Imidazóis/farmacologia , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/efeitos dos fármacos , Piridinas/farmacologia , RNA Interferente Pequeno/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Microb Pathog ; 50(5): 252-62, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21296653

RESUMO

The eukaryotic DNA recombination repair protein BRCA2 is functional in the parasitic protozoan Trypanosoma brucei. The mechanism of the involvement of BRCA2 in homologous recombination includes its interaction with the DNA recombinase proteins of the RAD51 family. BRCA2 is known to interact with RAD51 through its unique and essential BRC sequence motifs. T. brucei BRCA2 homolog (TbBRCA2) has fifteen repeating BRC motifs as compared to mammalian BRCA2 that has only eight. We report here our yeast 2-hybrid analysis studies on the interactions of TbBRCA2 BRC motifs with five different RAD51 paralogues of T. brucei. Our study revealed that a single BRC motif is sufficient to bind to these RAD51 paralogues. To test the possibility whether a single 44 amino acid long repeating unit of the TbBRCA2 BRC motif may be exploited as an inhibitor of T. brucei growth, we ectopically expressed this peptide segment in the procyclic form of the parasite and evaluated its effects on cell survival as well as the sensitivity of these cells to the DNA damaging agent methyl methane sulfonate (MMS). Expression of a single BRC motif led to MMS sensitivity and inhibited cellular proliferation in T. brucei.


Assuntos
Proteína BRCA2/metabolismo , Reparo do DNA , Proteínas de Protozoários/metabolismo , Rad51 Recombinase/metabolismo , Trypanosoma brucei brucei/fisiologia , Aptâmeros de Peptídeos/metabolismo , Proteína BRCA2/antagonistas & inibidores , Sobrevivência Celular , Ligação Proteica , Proteínas de Protozoários/antagonistas & inibidores , Técnicas do Sistema de Duplo-Híbrido
14.
Mol Cell Biochem ; 350(1-2): 47-57, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21165676

RESUMO

Leishmania is a group of parasitic protozoa that infect blood and tissue phagocytes including macrophages. We hypothesize that Leishmania is capable of establishing infection inside the macrophages because (a) they infect a subpopulation of macrophages; and (b) they "renovate" the macrophages before the establishment of infection. We found that only alternatively activated polarized M2 macrophages support Leishmania growth. Exposure of M2 macrophages to Leishmania promastigotes represses several selected RNA polymerase III (PolIII)-transcribed non-coding RNA (ncRNA) genes including those of 7SL RNA, vault RNA, and B2 RNA which have B-box element at their promoters. The B-box-binding transcription factor TFIIIC110 is down-regulated in Leishmania-exposed macrophages. Both the surface protease gp63 and the surface glycolipid LPG are required for the down-regulation of the ncRNAs in the M2 macrophages. We conclude that Leishmania surface gp63 collaborates with LPG to down-regulate TFIIIC110 in M2 macrophages to repress B-box containing ncRNA gene promoters.


Assuntos
Polaridade Celular , Leishmania/fisiologia , Macrófagos/metabolismo , Regiões Promotoras Genéticas , RNA não Traduzido/genética , Elementos Reguladores de Transcrição , Animais , Sequência de Bases , Polaridade Celular/fisiologia , Células Cultivadas , Regulação para Baixo/fisiologia , Regulação da Expressão Gênica , Humanos , Ativação de Macrófagos/genética , Ativação de Macrófagos/fisiologia , Macrófagos/fisiologia , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , RNA Citoplasmático Pequeno/química , RNA Citoplasmático Pequeno/genética , Elementos Reguladores de Transcrição/fisiologia , Partícula de Reconhecimento de Sinal/química , Partícula de Reconhecimento de Sinal/genética , Células U937
15.
Artigo em Inglês | MEDLINE | ID: mdl-32940190

RESUMO

The development of colorectal cancer (CRC) is a multistage process. The inflammation of the colon as in inflammatory bowel disease (IBD) such as ulcerative colitis (UC) or Crohn's disease (CD) is often regarded as the initial trigger for the development of inflammation-associated CRC. Many cytokines such as tumor necrosis factor alpha (TNF-α) and interleukins (ILs) are known to exert proinflammatory actions, and inflammation initiates or promotes tumorigenesis of various cancers, including CRC, through differential regulation of microRNAs (miRNAs/miRs). miRNAs can be oncogenic miRNAs (oncomiRs) or anti-oncomiRs/tumor suppressor miRNAs, and they play key roles during colorectal carcinogenesis. However, the functions and molecular mechanisms of regulation of miRNAs involved in inflammation-associated CRC are still anecdotal and largely unknown. Consolidating the published results and offering perspective solutions to circumvent CRC, the current review is focused on the role of miRNAs and their regulation in the development of CRC. We have also discussed the model systems adapted by researchers to delineate the role of miRNAs in inflammation-associated CRC.


Assuntos
Neoplasias Colorretais/genética , Doenças Inflamatórias Intestinais/complicações , MicroRNAs/fisiologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Colite/complicações , Colite/genética , Colite/patologia , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Inflamação/complicações , Inflamação/genética , Inflamação/patologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Transdução de Sinais/genética
16.
Artigo em Inglês | MEDLINE | ID: mdl-32901590

RESUMO

Chronic inflammation can lead to the development of many diseases, including cancer. Inflammatory bowel disease (IBD) that includes both ulcerative colitis (UC) and Crohnmp's disease (CD) are risk factors for the development of colorectal cancer (CRC). Many cytokines produced primarily by the gut immune cells either during or in response to localized inflammation in the colon and rectum are known to stimulate the complex interactions between the different cell types in the gut environment resulting in acute inflammation. Subsequently, chronic inflammation, together with genetic and epigenetic changes, have been shown to lead to the development and progression of CRC. Various cell types present in the colon, such as enterocytes, Paneth cells, goblet cells, and macrophages, express receptors for inflammatory cytokines and respond to tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), IL-6, and other cytokines. Among the several cytokines produced, TNF-α and IL-1ß are the key pro-inflammatory molecules that play critical roles in the development of CRC. The current review is intended to consolidate the published findings to focus on the role of pro-inflammatory cytokines, namely TNF-α and IL-1ß, on inflammation (and the altered immune response) in the gut, to better understand the development of CRC in IBD, using various experimental model systems, preclinical and clinical studies. Moreover, this review also highlights the current therapeutic strategies available (monotherapy and combination therapy) to alleviate the symptoms or treat inflammation-associated CRC by using monoclonal antibodies or aptamers to block pro-inflammatory molecules, inhibitors of tyrosine kinases in the inflammatory signaling cascade, competitive inhibitors of pro-inflammatory molecules, and the nucleic acid drugs like small activating RNAs (saRNAs) or microRNA (miRNA) mimics to activate tumor suppressor or repress oncogene/pro-inflammatory cytokine gene expression.


Assuntos
Neoplasias Colorretais/etiologia , Inflamação/complicações , Doenças Inflamatórias Intestinais/complicações , Animais , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Colite/complicações , Colite/patologia , Neoplasias Colorretais/patologia , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Inflamação/patologia , Doenças Inflamatórias Intestinais/patologia
17.
Mol Cancer ; 9: 50, 2010 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-20202217

RESUMO

BACKGROUND: BRCA2 gene expression is tightly regulated during the cell cycle in human breast cells. The expression of BRCA2 gene is silenced at the G0/G1 phase of cell growth and is de-silenced at the S/G2 phase. While studying the activity of BRCA2 gene promoter in breast cancer cells, we discovered that this promoter has bi-directional activity and the product of the reverse activity (a ZAR1-like protein, we named ZAR2) silences the forward promoter at the G0/G1 phase of the cell. Standard techniques like cell synchronization by serum starvation, flow cytometry, N-terminal or C-terminal FLAG epitope-tagged protein expression, immunofluorescence confocal microscopy, dual luciferase assay for promoter evaluation, and chromatin immunoprecipitation assay were employed during this study. RESULTS: Human BRCA2 gene promoter is active in both the forward and the reverse orientations. This promoter is 8-20 fold more active in the reverse orientation than in the forward orientation when the cells are in the non-dividing stage (G0/G1). When the cells are in the dividing state (S/G2), the forward activity of the promoter is 5-8 folds higher than the reverse activity. The reverse activity transcribes the ZAR2 mRNA with 966 nt coding sequence which codes for a 321 amino acid protein. ZAR2 has two C4 type zinc fingers at the carboxyl terminus. In the G0/G1 growth phase ZAR2 is predominantly located inside the nucleus of the breast cells, binds to the BRCA2 promoter and inhibits the expression of BRCA2. In the dividing cells, ZAR2 is trapped in the cytoplasm. CONCLUSIONS: BRCA2 gene promoter has bi-directional activity, expressing BRCA2 and a novel C4-type zinc finger containing transcription factor ZAR2. Subcellular location of ZAR2 and its expression from the reverse promoter of the BRCA2 gene are stringently regulated in a cell cycle dependent manner. ZAR2 binds to BRCA2/ZAR2 bi-directional promoter in vivo and is responsible, at least in part, for the silencing of BRCA2 gene expression in the G0/G1 phase in human breast cells.


Assuntos
Proteína BRCA2/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Regiões 5' não Traduzidas/genética , Animais , Proteína BRCA2/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Códon/genética , Sequência Conservada , Citosol/metabolismo , Éxons/genética , Feminino , Fase G1 , Técnicas de Silenciamento de Genes , Rearranjo Gênico/genética , Humanos , Íntrons/genética , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fase de Repouso do Ciclo Celular , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/metabolismo , Vertebrados/genética
18.
J Neurosci Res ; 88(15): 3386-98, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20857507

RESUMO

We examined the effects of 45-min hypoxia (FiO(2) 0.08; Hx) vs. normoxia (FiO(2) 0.21; Nx) on the ipsilateral (Ipsi) and contralateral (Ctrl) sides of the brain in neuronal glucose transporter isoform 3 (Glut3) heterozygous null mice (glut3(+/-)) and their wild-type littermates (WT), undergoing unilateral carotid artery ligation. Glut3(+/-) mice, under Nx, demonstrated a compensatory increase in blood-brain barrier/glial Glut1 protein concentration and a concomitant increase in neuronal nitric oxide synthase (nNOS) enzyme activity and Bax protein, with a decrease in procaspase 3 protein (P < 0.05 each). After Hx, reoxygenation in FiO(2) of 0.21 led to no comparable adaptive up-regulation of the ipsilateral brain Glut3 or Glut1 protein at 4 hr and Glut1 at 24 hr in glut3(+/-) vs. WT. These brain Glut changes in glut3(+/-) but not WT mice were associated with an increase in proapoptotic Bax protein and caspase-3 enzyme activity (P < 0.01 each) and a decline in the antiapoptotic Bcl-2 and procaspase-3 proteins (P < 0.05 each). Glut3(+/-) mice after Hx demonstrated TUNEL-positive neurons with nuclear pyknosis in most ipsilateral (hypoxic-ischemia) brain regions. A subset (∼55%) of glut3(+/-) mice developed spontaneous seizures after hypoxic-ischemia, confirmed by electroencephalography, but the WT mice remained seizure-free. Pentylenetetrazole testing demonstrated an increased occurrence of longer lasting clinical seizures at a lower threshold in glut3(+/-) vs. WT mice, with no detectable differences in monamine neurotransmitters. We conclude that hypoxic-ischemic brain injury in glut3(+/-) mice exacerbates cellular apoptosis and necrosis and precipitates spontaneous seizures.


Assuntos
Apoptose/fisiologia , Transportador de Glucose Tipo 3/metabolismo , Hipóxia-Isquemia Encefálica/fisiopatologia , Neurônios/patologia , Convulsões/fisiopatologia , Animais , Western Blotting , Cromatografia Líquida de Alta Pressão , Feminino , Imunofluorescência , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 3/genética , Heterozigoto , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neurônios/metabolismo , Fenótipo , Convulsões/genética , Convulsões/metabolismo
19.
Mol Biol Rep ; 37(3): 1221-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19277896

RESUMO

SNAI1P, a protein coded by a retrogene, is a member of the SNAI family of E2-box binding transcriptional repressors. To evaluate whether the mode of action of SNAI1P is similar to those of the other predominant members of the SNAI family, we studied its action on human claudin 7 (CLDN7) gene promoter which has seven E2-boxes. We over-expressed FLAG-tagged SNAI1P in MCF7 and MDA-MB-468 cells. SNAI1P inhibited the expression of CLDN7 in these recombinant cells. SNAI1P also inhibited cloned CLDN7 gene promoter activity in human breast cancer cells. ChIP assays revealed that SNAI1P is recruited on the CLDN7 gene promoter along with the co-repressor CtBP1 and the effector HDAC1. Treatment of the cells with trichostatin A, an inhibitor of HDAC1, abrogated the repressor activity of SNAI1P. These data suggest that SNAI1P inhibits CLDN7 gene promoter epigenetically in breast cancer cells through chromatin remodeling.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , Epigênese Genética , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina/genética , Imunoprecipitação da Cromatina , Claudinas , Regulação da Expressão Gênica/genética , Humanos , Ácidos Hidroxâmicos , Proteínas de Membrana/genética , Regiões Promotoras Genéticas/genética , Fatores de Transcrição da Família Snail
20.
Cancer Res ; 67(1): 289-99, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17210710

RESUMO

Nitric oxide (NO) in nanomolar (nmol/L) concentrations is consistently detected in tumor microenvironment and has been found to promote tumorigenesis. The mechanism by which NO enhances tumor progression is largely unknown. In this study, we investigated the possible mechanisms and identified cellular targets by which NO increases proliferation of human breast cancer cell lines MDA-MB-231 and MCF-7. DETA-NONOate, a long acting NO donor, with a half-life of 20 h, was used. We found that NO (nmol/L) dramatically increased total protein synthesis in MDA-MB-231 and MCF-7 and also increased cell proliferation. NO specifically increased the translation of cyclin D1 and ornithine decarboxylase (ODC) without altering their mRNA levels or half-lives. Critical components in the translational machinery, such as phosphorylated mammalian target of rapamycin (mTOR) and its downstream targets, phosphorylated eukaryotic translation initiation factor and p70 S6 kinase, were up-regulated following NO treatment, and inhibition of mTOR with rapamycin attenuated NO induced increase of cyclin D1 and ODC. Activation of translational machinery was mediated by NO-induced up-regulation of the Raf/mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase/ERK (Raf/MEK/ERK) and phosphatidylinositol 3-kinase (PI-3 kinase)/Akt signaling pathways. Up-regulation of the Raf/MEK/ERK and PI-3 kinase/Akt pathways by NO was found to be mediated by activation of Ras, which was cyclic guanosine 3',5'-monophosphate independent. Furthermore, inactivation of Ras by farnesyl transferase inhibitor or K-Ras small interfering RNA attenuated NO-induced increase in proliferation signaling and cyclin D1 and ODC translation, further confirming the involvement of Ras activation during NO-induced cell proliferation.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fator de Iniciação 4E em Eucariotos/metabolismo , Óxido Nítrico/farmacologia , Proteínas Quinases/metabolismo , Alquil e Aril Transferases/antagonistas & inibidores , Neoplasias da Mama/genética , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , GMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Humanos , Sistema de Sinalização das MAP Quinases , Compostos Nitrosos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR , Quinases raf/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa