Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Free Radic Res ; 54(11-12): 906-917, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32336239

RESUMO

Recent studies report that nuclear factor-erythroid-2-related factor 2 (Nrf2) facilitates tumor progression through metabolic reprogramming in cancer cells. However, the molecular mechanism underlying the oncogenic functions of Nrf2 is not yet well understood. Some of the pentose phosphate pathway (PPP) enzymes are considered to play a role in the cancer progression. The present study was intended to explore the potential role of phosphogluconate dehydrogenase (PGD), one of the PPP enzymes, in the proliferation and migration of human hepatoma HepG2 cells. Genetic ablation of Nrf2 attenuated the expression of PGD at both transcriptional and translational levels. Notably, Nrf2 regulates the transcription of PGD through direct binding to the antioxidant response element in its promoter region. Nrf2 overexpression in HepG2 cells led to increased proliferation, survival, and migration, and these events were suppressed by silencing PGD. Interestingly, knockdown of the gene encoding this enzyme not only attenuated the proliferation and clonogenicity of HepG2 cells but also downregulated the expression of Nrf2. Thus, there seems to exist a positive feedback loop between Nrf2 and PGD which is exploited by hepatoma cells for their proliferation and survival. Treatment of HepG2 cells with ribulose-5-phosphate, a catalytic product of PGD, gave rise to a concentration-dependent upregulation of Nrf2. Collectively, the current study shows that Nrf2 promotes hepatoma cell growth and progression, partly through induction of PGD transcription.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fosfogluconato Desidrogenase/metabolismo , Proliferação de Células , Células Hep G2 , Humanos , Transfecção
2.
BMB Rep ; 52(11): 659-664, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31072447

RESUMO

Vav1 is a Rho/Rac guanine nucleotide exchange factor primarily expressed in hematopoietic cells. In this study, we investigated the potential role of Vav1 in osteoclast (OC) differentiation by comparing the ability of bone marrow mononuclear cells (BMMCs) obtained from Vav1-deficient (Vav1-/-) and wild-type (WT) mice to differentiate into mature OCs upon stimulation with macrophage colony stimulating factor and receptor activator of nuclear kappa B ligand in vitro. Our results suggested that Vav1 deficiency promoted the differentiation of BMMCs into OCs, as indicated by the increased expression of tartrate-resistant acid phosphatase, cathepsin K, and calcitonin receptor. Therefore, Vav1 may play a negative role in OC differentiation. This hypothesis was supported by the observation of more OCs in the femurs of Vav1-/- mice than in WT mice. Furthermore, the bone status of Vav1-/- mice was analyzed in situ and the femurs of Vav1-/- mice appeared abnormal, with poor bone density and fewer number of trabeculae. In addition, Vav1-deficient OCs showed stronger adhesion to vitronectin, an αvß3 integrin ligand important in bone resorption. Thus, Vav1 may inhibit OC differentiation and protect against bone resorption. [BMB Reports 2019; 52(11): 659-664].


Assuntos
Osteoclastos/metabolismo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Animais , Células da Medula Óssea/metabolismo , Reabsorção Óssea/metabolismo , Osso e Ossos/metabolismo , Diferenciação Celular/fisiologia , Integrina alfaVbeta3/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/citologia , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/fisiologia , Ligante RANK/metabolismo , Ligante RANK/fisiologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Fosfatase Ácida Resistente a Tartarato
3.
Free Radic Res ; 52(11-12): 1336-1347, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30238818

RESUMO

Cancer stem cells (CSCs) constitute a subpopulation of transformed cells that possess intrinsic ability to undergo selfrenewal and differentiation, which drive tumour resistance and cancer recurrence. It has been reported that CSCs possess enhanced protection against oxidative stress induced by reactive oxygen species compared with nonstem-like cancer cells. In the present work, we investigated the role of heme oxygenase-1 (HO-1), a representative antioxidant enzyme, on the stemness and selfrenewal of human breast CSCs. We found that pharmacologic or genetic inhibition of HO-1 attenuated the sphere formation, whereas HO-1 inducers enhanced the number and the size of tumourspheres in breast CSCs. Carbon monoxide (CO) is endogenously generated as a consequence of degradation of heme by HO-1. The proportion of populations of CD44+/CD24- cells retaining CSC properties was increased in MDA-MB-231 cells treated with a CO-releasing molecule (CORM-2). Following CORM-2 treatment, the expression of Notch-1 and related genes Jagged-1 and Hes1 was increased, which was accompanied by the mammosphere formation. Taken together, these findings suggest that HO-1-derived CO production stimulates the formation of mammospheres in breast cancer cells through activation of Notch-1 signalling.


Assuntos
Neoplasias da Mama/metabolismo , Monóxido de Carbono/metabolismo , Heme Oxigenase-1/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptor Notch1/metabolismo , Neoplasias da Mama/patologia , Monóxido de Carbono/química , Feminino , Humanos , Células-Tronco Neoplásicas/patologia , Transdução de Sinais , Células Tumorais Cultivadas
4.
FASEB J ; 32(10): 5312-5325, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29718706

RESUMO

A key event required for effective resolution of inflammation is efferocytosis, which is defined as phagocytic removal of apoptotic cells mostly by macrophages acquiring an alternatively activated phenotype (M2). c-Myc has been reported to play a role in alternative activation of human macrophages and is proposed as one of the M2 macrophage markers. We found that M2-like peritoneal macrophages from zymosan A-treated mice exhibited a marked accumulation of Myc-nick, a truncated protein generated by a Calpain-mediated proteolytic cleavage of full-length c-Myc. Further, ectopic expression of Myc-nick in murine bone marrow-derived macrophages promoted the M2 polarization and, consequently, enhanced their efferocytic capability. Notably, Myc-nick-induced efferocytosis was found to be tightly associated with α-tubulin acetylation by K acetyltransferase 2a (Kat2a/Gcn5) activity. These findings suggest Myc-nick as a novel proresolving mediator that has a fundamental function in maintaining homeostasis under inflammatory conditions.-Zhong, X., Lee, H.-N., Kim, S. H., Park, S.-A., Kim, W., Cha, Y.-N., Surh, Y.-J. Myc-nick promotes efferocytosis through M2 macrophage polarization during resolution of inflammation.


Assuntos
Células da Medula Óssea/imunologia , Macrófagos Peritoneais/imunologia , Proteínas Proto-Oncogênicas c-myc/imunologia , Acetilação , Animais , Antígenos de Diferenciação/imunologia , Células da Medula Óssea/patologia , Histona Acetiltransferases/imunologia , Inflamação/imunologia , Inflamação/patologia , Macrófagos Peritoneais/patologia , Camundongos , Tubulina (Proteína)/imunologia , Fatores de Transcrição de p300-CBP/imunologia
5.
Sci Rep ; 8(1): 6409, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29686295

RESUMO

Signal transducer and activator of transcription 3 (STAT3) is a transcription factor that is latent but constitutively activated in many types of cancers. It is well known that STAT3 plays a key role in inflammation-associated tumorigenesis. Curcumin is an anti-inflammatory natural compound isolated from the turmeric (Curcuma longa L., Zingiberaceae) that has been extensively used in a traditional medicine over the centuries. In the present study, we have found that curcumin inhibits STAT3 signaling that is persistently overactivated in H-Ras transformed breast epithelial cells (H-Ras MCF10A). Specific cysteine residues present in STAT3 appear to be critical for the activity as well as conformation of this transcription factor. We identified the cysteine residue 259 of STAT3 as a putative site for curcumin binding. Site-directed mutation of this cysteine residue abolished curcumin-induced inactivation of STAT3 and apoptosis in H-Ras MCF10A cells. The α,ß-unsaturated carbonyl moiety of curcumin appears to be essential in its binding to STAT3 in H-Ras MCF10A cells. Tetrahydrocurcumin that lacks such electrophilic moiety failed to interact with STAT3 and to induce apoptosis in the same cell line. Taken together, our findings suggest that curcumin can abrogate aberrant activation of STAT3 through direct interaction, thereby inhibiting STAT3-mediated mammary carcinogenesis.


Assuntos
Apoptose/efeitos dos fármacos , Curcumina/metabolismo , Curcumina/farmacologia , Cisteína/metabolismo , Genes ras , Glândulas Mamárias Humanas/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Linhagem Celular Transformada , Curcumina/análogos & derivados , DNA/metabolismo , Dimerização , Humanos , Glândulas Mamárias Humanas/patologia , Fator de Transcrição STAT3/química , Compostos de Sulfidrila/metabolismo , Transcrição Gênica
6.
Cell Immunol ; 327: 36-46, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29477410

RESUMO

Phagocytosis of pathogens by macrophages is crucial for the successful resolution of inflammation induced by microbial infection. Taurine chloramine (TauCl), an endogenous anti-inflammatory and antioxidative substance, is produced by reaction between taurine and hypochlorous acid by myeloperoxidase activity in neutrophils under inflammatory conditions. In the present study, we investigated the effect of TauCl on resolution of acute inflammation caused by fungal infection using a zymosan A-induced murine peritonitis model. TauCl administration reduced the number of the total peritoneal leukocytes, while it increased the number of peritoneal monocytes. Furthermore, TauCl promoted clearance of pathogens remaining in the inflammatory environment by macrophages. When the macrophages isolated from thioglycollate-treated mice were treated with TauCl, their phagocytic capability was enhanced. In the murine macrophage-like RAW264.7 cells treated with TauCl, the proportion of macrophages clearing the zymosan A particles was also increased. TauCl administration resulted in elevated expression of heme oxygenase-1 (HO-1) in the peritoneal macrophages. Pharmacologic inhibition of HO-1 activity or knockdown of HO-1 in the murine macrophage RAW264.7 cells abolished the TauCl-induced phagocytosis, whereas the overexpression of HO-1 augmented the phagocytic ability of macrophages. Moreover, peritoneal macrophages isolated from HO-1 null mice failed to mediate TauCl-induced phagocytosis. Our results suggest that TauCl potentiates phagocytic activity of macrophages through upregulation of HO-1 expression.


Assuntos
Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/fisiologia , Taurina/análogos & derivados , Animais , Antioxidantes , Inflamação , Macrófagos/fisiologia , Macrófagos Peritoneais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/induzido quimicamente , Peritonite/fisiopatologia , Fagócitos , Fagocitose/fisiologia , Células RAW 264.7 , Taurina/metabolismo , Taurina/farmacologia , Regulação para Cima , Zimosan/farmacologia
7.
FASEB J ; 32(4): 2246-2257, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29247123

RESUMO

Resolution of inflammation that occurs after microbial infection or tissue damage is an important physiologic process in maintaining or restoring host homeostasis. Taurine chloramine (TauCl) is formed by a reaction between taurine and hypochlorite in leukocytes, and it is especially abundant in activated neutrophils that encounter an oxidative burst. As neutrophils undergo apoptosis, TauCl is released to the extracellular matrix at the inflamed sites, thereby affecting coexisting macrophages in the inflammatory microenvironment. In this study, we investigated the role of TauCl in phagocytosis by macrophages during resolution of fungal infection-induced inflammation. We found that exogenous TauCl substantially increased the phagocytic efficiency of macrophages through up-regulation of dectin-1, a receptor for fungal ß-1,3-glucans, which is present on the membrane of macrophages. Our previous studies demonstrated the induction of heme oxygenase-1 (HO-1) expression in murine peritoneal macrophages treated with TauCl. In the present study, knocking out HO-1 or pharmacologic inhibition of HO-1 with zinc protoporphyrin IX attenuated the TauCl-induced expression of dectin-1 and subsequent phagocytosis. Furthermore, carbon monoxide (CO), a by-product of the HO-1-catalyzed reaction, induced expression of dectin-1 and potentiated phagocytic capability of the macrophages, which appeared to be mediated through up-regulation of peroxisome proliferator-activated receptor γ. Taken together, induction of HO-1 expression and subsequent CO production by TauCl are essential for phagocytosis of fungi by macrophages. Our results suggest that TauCl has important roles in host defense against fungal infection and has therapeutic potential in the management of inflammatory diseases.-Kim, S. H., Zhong, X., Kim, W., Kim, K., Suh, Y.-G., Kim, C., Joe, Y., Chung, H. T., Cha, Y.-N., Surh, Y.-J. Taurine chloramine potentiates phagocytic activity of peritoneal macrophages through up-regulation of dectin-1 mediated by heme oxygenase-1-derived carbon monoxide.


Assuntos
Inibidores Enzimáticos/farmacologia , Lectinas Tipo C/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Fagocitose , Taurina/análogos & derivados , Regulação para Cima , Animais , Candida albicans/patogenicidade , Monóxido de Carbono/metabolismo , Células Cultivadas , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Lectinas Tipo C/genética , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama , Taurina/farmacologia
8.
Sci Rep ; 7(1): 15918, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29162839

RESUMO

During the metastatic phase, cancer cells require the dissolution of cadherin-mediated cell-cell adhesion and a dramatic re-organization of the cytoskeleton through epithelial-mesenchymal transition (EMT), thereby acquiring migratory and invasive capabilities. In most tumors, EMT is accompanied by hypoxia. However, the intracellular signaling molecule that mediates hypoxia-induced EMT remained overlooked. By utilizing the microarray database system of the Cancer Genome Atlas, we identified ubiquitin-specific protease 47 (USP47), a deubiquitinating enzyme, as a potential mediator of hypoxia-induced EMT. Immunofluorescence staining of human colorectal tissue microarrays revealed that USP47 is overexpressed in colorectal adenocarcinoma tissues compared with normal adjacent tissues. The expression of USP47 was found to be elevated in three different human colorectal cancer cell lines. The enhancement of USP47 in colorectal cancer cells under hypoxic conditions induced the disassembly of E-cadherin and promoted EMT through deubiquitination of Snail. Silencing of USP47 accelerated the proteasomal degradation of Snail and inhibited EMT. Notably, hypoxia-induced USP47 upregulation was mediated by Sox9. These results demonstrate, for the first time, the role for USP47, as a novel target of Sox9, in the regulation of EMT and metastasis of colorectal cancer cells.


Assuntos
Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Ubiquitina Tiolesterase/metabolismo , Animais , Hipóxia Celular/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Biológicos , Invasividade Neoplásica , Metástase Neoplásica , Regiões Promotoras Genéticas/genética , Estabilidade Proteica , Proteases Específicas de Ubiquitina , Ubiquitinação , Regulação para Cima/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cancer Res ; 77(18): 4797-4808, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28655791

RESUMO

Nrf2, a master regulator of oxidative stress, is considered a prominent target for prevention of hepatocellular carcinoma (HCC), one of the leading causes of cancer-related deaths worldwide. Here we report that Nrf2-deficient mice resisted diethylnitrosamine (DEN)-induced hepatocarcinogenesis without affecting P450-mediated metabolic activation of DEN. Nrf2 expression, nuclear translocation, and transcriptional activity were enhanced in liver tumors. Overactivated Nrf2 was required for hepatoma growth in DEN-induced HCC. Following DEN treatment, Nrf2 genetic disruption reduced expression of pentose phosphate pathway-related enzymes, the depletion of which has been associated with an amelioration of HCC incidence. Conversely, enhanced Nrf2 activity was attributable to alterations in the ability to bind its endogenous inhibitor Keap1. Our findings provide a mechanistic rationale for Nrf2 blockade to prevent and possibly treat liver cancer. Cancer Res; 77(18); 4797-808. ©2017 AACR.


Assuntos
Dietilnitrosamina/toxicidade , Neoplasias Hepáticas Experimentais/patologia , Subunidade p45 do Fator de Transcrição NF-E2/fisiologia , Alquilantes/toxicidade , Animais , Apoptose , Proliferação de Células , Feminino , Humanos , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênicos/toxicidade , Transdução de Sinais , Células Tumorais Cultivadas
10.
Antioxid Redox Signal ; 27(17): 1412-1431, 2017 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-28398824

RESUMO

AIMS: 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) has been shown to rescue cells from inflammatory insults and to participate in the resolution of acute inflammation. In this study, we investigated molecular mechanisms underlying proresolving effects of 15d-PGJ2. RESULTS: 15d-PGJ2 injected into the peritoneum of mice facilitated the resolution of zymosan A-induced peritonitis. 15d-PGJ2 administration reduced the number of total leukocytes and attenuated polymorphonuclear leukocyte infiltration. Furthermore, 15d-PGJ2 increased the proportion of macrophages engulfing apoptotic neutrophils, a process called efferocytosis. In addition, when the thioglycollate-elicited mouse peritoneal macrophages were stimulated with 15d-PGJ2, their efferocytic activity was amplified. In another experiment, RAW264.7 murine macrophages exposed to 15d-PGJ2 conducted phagocytic clearance of apoptotic cells to a greater extent than the control cells. Under these conditions, expression of CD36 and heme oxygenase-1 (HO-1) was enhanced along with increased accumulation of the nuclear factor E2-related factor 2 (Nrf2) in the nucleus. Knockdown of Nrf2 abolished 15d-PGJ2-induced expression of CD36 and HO-1, and silencing of CD36 and HO-1 attenuated 15d-PGJ2-induced efferocytosis. Moreover, peritoneal macrophages isolated from Nrf2-null mice failed to upregulate 15d-PGJ2-induced expression of CD36 and HO-1 and to mediate efferocytosis. Unlike 15d-PGJ2, its nonelectrophilic analog 9,10-dihydro-15d-PGJ2 lacking the α,ß-unsaturated carbonyl group could not induce CD36 expression and efferocytosis. INNOVATION: 15d-PGJ2, as one of the terminal products of cyclooxygenase-2, exerts proresolving effects through induction of efferocytosis. The results of this study suggest that 15d-PGJ2 possesses a therapeutic value in the management of inflammatory disorders. CONCLUSION: 15d-PGJ2 facilitates resolution of inflammation by inducing Nrf2-induced expression of CD36 and HO-1 in macrophages. Antioxid. Redox Signal. 27, 1412-1431.


Assuntos
Antígenos CD36/metabolismo , Heme Oxigenase-1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Peritonite/tratamento farmacológico , Prostaglandina D2/análogos & derivados , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células Jurkat , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Peritonite/induzido quimicamente , Fagocitose , Prostaglandina D2/administração & dosagem , Prostaglandina D2/farmacologia , Células RAW 264.7 , Zimosan/efeitos adversos
11.
Free Radic Res ; 50(10): 1140-1152, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27598034

RESUMO

15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), a representative J-series cyclopentenone prostaglandin, has biphasic roles in cell proliferation and apoptosis. Hypoxia inducible factor-1 (HIF-1) regulates expression of various genes involved in tumor growth and angiogenesis. In the present study, treatment of human breast cancer (MCF-7) cells with 15d-PGJ2 resulted in the accumulation of the α-subunit of HIF-1. Pretreatment with zinc protoporphyrin IX, a pharmacological inhibitor of heme oxygenase-1 (HO-1), as well as siRNA knockdown of HO-1 gene in MCF-7 cells attenuated 15d-PGJ2-mediated HIF-1α accumulation. 15d-PGJ2 treatment increased intracellular production of reactive oxygen species (ROS), which was mediated by HO-1 induction. Preincubation of MCF-7 cells with trolox, a water-soluble form of vitamin E, attenuated 15d-PGJ2-induced HIF-1α expression although HO-1 expression was unchanged. This finding suggests that ROS accumulated as a consequence of HO-1 up-regulation can enhance HIF-1α expression in MCF-7 cells treated with 15d-PGJ2. Alternatively, 15d-PGJ2 was found to covalently bind to HIF-1α prolyl-4-hydroxylase 2 (PHD2) in MCF-7 cells, which hampers the proline hydroxylation of HIF-1α, thereby disrupting ubiquitin-dependent proteasomal degradation of this transcription factor. Pretreatment with thiol reducing agents blunted 15d-PGJ2-induced HIF-1α stabilization, indicative of a cysteine residue as a direct target of 15d-PGJ2. Molecular docking analysis suggests that 15d-PGJ2 preferentially binds to PHD2 in the vicinity of the Cys201 residue based on binding energies and carbon-sulfur distances. In summary, 15d-PGJ2 stabilizes HIF-1α in MCF-7 cells through HO-1 induction with subsequent ROS generation and also through direct modification of PHD2.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Prolil Hidroxilases/metabolismo , Heme Oxigenase-1/metabolismo , Humanos
12.
Helicobacter ; 21(5): 405-16, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26910614

RESUMO

BACKGROUND: Recent studies have shown that Helicobacter pylori (H. pylori) activates signal transducer and activator of transcription 3 (STAT3) that plays an important role in gastric carcinogenesis. However, the molecular mechanism underlying H. pylori-mediated STAT3 activation is still not fully understood. In this study, we investigated H. pylori-induced activation of STAT3 signaling in AGS human gastric cancer cells and the underlying mechanism. MATERIALS AND METHODS: AGS cells were cocultured with H. pylori, and STAT3 activation was assessed by Western blot analysis, electrophoretic mobility shift assay and immunocytochemistry. To demonstrate the involvement of reactive oxygen species (ROS) in H. pylori-activated STAT3 signaling, the antioxidant N-acetylcysteine was utilized. The expression and production of interleukin-6 (IL-6) were measured by reverse-transcription polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA), respectively. The interaction between IL-6 and IL-6 receptor (IL-6R) was determined by the immunoprecipitation assay. RESULTS: H. pylori activates STAT3 as evidenced by increases in phosphorylation on Tyr(705) , nuclear localization, DNA binding and transcriptional activity of this transcription factor. The nuclear translocation of STAT3 was also observed in H. pylori-inoculated mouse stomach. In the subsequent study, we found that H. pylori-induced STAT3 phosphorylation was dependent on IL-6. Notably, the increased IL-6 expression and the IL-6 and IL-6R binding were mediated by ROS produced as a consequence of H. pylori infection. CONCLUSIONS: H. pylori-induced STAT3 activation is mediated, at least in part, through ROS-induced upregulation of IL-6 expression. These findings provide a novel molecular mechanism responsible for H. pylori-induced gastritis and gastric carcinogenesis.


Assuntos
Helicobacter pylori/imunologia , Interleucina-6/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Western Blotting , Linhagem Celular Tumoral , Técnicas de Cocultura , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Imuno-Histoquímica , Imunoprecipitação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ativação Transcricional
13.
PLoS One ; 11(1): e0147038, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26784107

RESUMO

Peptidyl prolyl isomerase (PIN1) regulates the functional activity of a subset of phosphoproteins through binding to phosphorylated Ser/Thr-Pro motifs and subsequently isomerization of the phosphorylated bonds. Interestingly, PIN1 is overexpressed in many types of malignancies including breast, prostate, lung and colon cancers. However, its oncogenic functions have not been fully elucidated. Here, we report that PIN1 directly interacts with hypoxia-inducible factor (HIF)-1α in human colon cancer (HCT116) cells. PIN1 binding to HIF-1α occurred in a phosphorylation-dependent manner. We also found that PIN1 interacted with HIF-1α at both exogenous and endogenous levels. Notably, PIN1 binding stabilized the HIF-1α protein, given that their levels were significantly increased under hypoxic conditions. The stabilization of HIF-1α resulted in increased transcriptional activity, consequently upregulating expression of vascular endothelial growth factor, a major contributor to angiogenesis. Silencing of PIN1 or pharmacologic inhibition of its activity abrogated the angiogenesis. By utilizing a bioluminescence imaging technique, we were able to demonstrate that PIN1 inhibition dramatically reduced the tumor volume in a subcutaneous mouse xenograft model and angiogenesis as well as hypoxia-induced transcriptional activity of HIF-1α. These results suggest that PIN1 interacting with HIF-1α is a potential cancer chemopreventive and therapeutic target.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/química , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Experimentais/irrigação sanguínea , Neovascularização Patológica/metabolismo , Peptidilprolil Isomerase/metabolismo , Animais , Hipóxia Celular , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HEK293 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Peptidilprolil Isomerase de Interação com NIMA , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neovascularização Patológica/genética , Fosforilação , Estabilidade Proteica , Fator A de Crescimento do Endotélio Vascular/genética
14.
Mol Carcinog ; 55(12): 2236-2246, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26808296

RESUMO

Helicobacter pylori (H. pylori) infection has been known to be implicated in human gastric carcinogenesis. Snail, the zinc-finger transcription factor known as a key inducer of changes in the cell shape and morphogenetic movement, is aberrantly overexpressed and correlates with lymph node metastasis in gastric cancer. In the present study, we investigated whether H. pylori could induce Snail activation to provoke these changes. Using a cell scatter assay, we noticed that human gastric cancer AGS cells infected with H. pylori underwent morphological changes as well as disruption of cell-cell interaction, which was then reversed by silencing of Snail by use of small interfering RNA (siRNA). In addition, infection with H. pylori resulted in an increased intracellular level of Snail in gastric cancer cells, which was abrogated in the presence of U0126 and LY294002, inhibitors of MEK/Erk and PI3K/Akt pathways, respectively. Cycloheximide pulse-chase experiments coupled with immunocytochemical analysis revealed that the induction of Snail by H. pylori was regulated at multiple levels, including increased transcription of Snail mRNA, inhibition of protein degradation, and enhancement of nuclear translocation of Snail. Pre-treatment of AGS cells with N-acetylcysteine, a well-known reactive oxygen species (ROS) scavenger, attenuated the H. pylori-induced activation of Erk, its binding to Snail promoter, inactivation of GSK-3ß, and accumulation of Snail. Collectively, these findings suggest that the upregulation of Snail expression induced by H. pylori and transformation to a spindle-like shape as a consequence in gastric cancer cells are attributable to ROS-mediated activation of Erk and the inhibition of GSK-3ß signaling. © 2016 Wiley Periodicals, Inc.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Infecções por Helicobacter/complicações , Helicobacter pylori/fisiologia , Sistema de Sinalização das MAP Quinases , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição da Família Snail/genética , Neoplasias Gástricas/virologia , Regulação para Cima , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/virologia , Humanos , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
15.
Antioxid Redox Signal ; 23(2): 163-77, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25816687

RESUMO

AIMS: To examine the pro-resolving effects of taurine chloramine (TauCl). RESULTS: TauCl injected into the peritoneum of mice enhanced the resolution of zymosan A-induced peritonitis. Furthermore, when the macrophages obtained from peritoneal exudates were treated with TauCl, their efferocytic ability was elevated. In the murine macrophage-like RAW264.7 cells exposed to TauCl, the proportion of macrophages engulfing the apoptotic neutrophils was also increased. In these macrophages treated with TauCl, expression of heme oxygenase-1 (HO-1) was elevated along with increased nuclear translocation of the nuclear factor E2-related factor 2 (Nrf2). TauCl binds directly to Kelch-like ECH association protein 1 (Keap1), which appears to retard the Keap1-driven degradation of Nrf2. This results in stabilization and enhanced nuclear translocation of Nrf2 and upregulation of HO-1 expression. TauCl, when treated to peritoneal macrophages isolated from either Nrf2 or HO-1 wild-type mice, stimulated efferocytosis (phagocytic engulfment of apoptotic neutrophils by macrophages), but not in the macrophages from Nrf2 or HO-1 knockout mice. Furthermore, transcriptional expression of some scavenger receptors recognizing the phosphatidylserines exposed on the surface of apoptotic cells was increased in RAW264.7 cells treated with TauCl. Pharmacologic inhibition of HO-1 activity or knockdown of HO-1 gene in RAW264.7 cells abolished the TauCl-induced efferocytosis, whereas both overexpression of HO-1 and treatment with carbon monoxide (CO), the product of HO, potentiated the efferocytic activity of macrophages. INNOVATION: This work provides the first evidence that TauCl stimulates efferocytosis by macrophages. The results of this study suggest the therapeutic potential of TauCl in the management of inflammatory disorders. CONCLUSION: TauCl can facilitate resolution of inflammation by increasing the efferocytic activity of macrophages through Nrf2-mediated HO-1 upregulation and subsequent production of CO.


Assuntos
Monóxido de Carbono/metabolismo , Heme Oxigenase-1/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos/metabolismo , Fagocitose/efeitos dos fármacos , Taurina/análogos & derivados , Regulação para Cima/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Células Jurkat , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Taurina/farmacologia , Zimosan/metabolismo
16.
Cancer Lett ; 351(1): 41-9, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-24747121

RESUMO

Sulforaphane, an isothiocyanate present in cruciferous vegetables, has been reported to possess anti-inflammatory and cancer chemopreventive properties. However, the molecular mechanisms by which sulforaphane suppresses inflammation and carcinogenesis are yet to be fully elucidated. Since the aberrant expression of cyclooxygenase-2 (COX-2) links inflammation and cancer, the present study was aimed to elucidate the mechanisms by which sulforaphane modulates COX-2 overexpression in human mammary epithelial (MCF-10A) cells stimulated with a prototypic tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA). Treatment of MCF-10A cells with sulforaphane significantly inhibited TPA-induced expression of COX-2 protein and its mRNA transcript. Transient transfection of cells with deletion mutant constructs of COX-2 promoter revealed that the transcription factor nuclear factor-kappaB (NF-κB) plays a key role in TPA-induced COX-2 expression in MCF-10A cells. Pretreatment with sulforaphane significantly attenuated nuclear localization, DNA binding and the transcriptional activity of NF-κB through inhibition of phosphorylation and subsequent degradation of IκBα in MCF-10A cells stimulated with TPA. Sulforaphane also attenuated TPA-induced activation of IκB kinases (IKK), NF-κB-activating kinase (NAK) and extracellular signal-regulated kinase-1/2 (ERK1/2). Pharmacological inhibition of IKK or transient transfection of cells with dominant-negative mutant forms of this kinase abrogated TPA-induced NF-κB activation and COX-2 expression. In addition, the blockade of ERK1/2 activation negated the catalytic activity of IKKα, but not that of IKKß, whereas silencing NAK by specific siRNA abrogated the IKKß activity in TPA-treated cells. Taken together, sulforaphane inhibits TPA-induced NF-κB activation and COX-2 expression in MCF-10A cells by blocking two distinct signaling pathways mediated by ERK1/2-IKKα and NAK-IKKß.


Assuntos
Anticarcinógenos/farmacologia , Ciclo-Oxigenase 2/metabolismo , Células Epiteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase I-kappa B/metabolismo , Isotiocianatos/farmacologia , NF-kappa B/metabolismo , Neoplasias da Mama/prevenção & controle , Linhagem Celular , Ciclo-Oxigenase 2/genética , Ensaios de Seleção de Medicamentos Antitumorais , Células Epiteliais/enzimologia , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases , Glândulas Mamárias Humanas/patologia , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Sulfóxidos , Acetato de Tetradecanoilforbol/farmacologia
17.
PLoS One ; 9(1): e85984, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489685

RESUMO

Diallyl sulfide, diallyl disulfide, and daillyl trisulfide (DATS) are major volatile components of garlic oil. In this study, we assessed their relative potency in inducing antioxidant enzyme expression. Among the three organosulfur compounds, DATS was found to be most potent in inducing heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO1) in human gastric epithelial (AGS) cells. Furthermore, DATS administration by gavage increased the expression of HO-1 and NQO1 in C57BL/6 mouse stomach. Treatment with DATS increased the accumulation of nuclear factor-erythroid-2-related factor-2 (Nrf2) in the nucleus of cultured AGS cells and in mouse stomach in vivo. The DATS-induced expression of HO-1 and NQO1 was abrogated in the cells transiently transfected with Nrf2-siRNA or in the embryonic fibroblasts from Nrf2-null mice, indicating that Nrf2 is a key mediator of the cytoprotective effects of DATS. Pretreatment of AGS cells with N-acetylcysteine or dithiothreitol attenuated DATS-induced nuclear localization of Nrf2 and the expression of HO-1 and NQO1. Cysteine-151, -273 and -288 of Kelch-like ECH-associated protein-1 (Keap1), a cytosolic repressor of Nrf2, have been considered to act as a redox sensor and play a role in Nrf2 activation. To determine whether DATS could inactivate Keap1 through thiol modification, we established cell lines constitutively expressing wild type-Keap1 or three different mutant constructs in which cysteine-151, -273, or -288 of Keap1 was replaced with serine by retroviral gene transfer. DATS failed to activate Nrf2, and to induce expression of HO-1 and NQO1 only in Keap1-C288S mutant cells. LC-ESI-MS/MS analysis of recombinant Keap1 treated with DATS revealed that the peptide fragment containing Cys288 gained a molecular mass of 72.1 Da equivalent to the molecular weight of mono-allyl mono-sulfide. Taken together, these findings suggest that DATS may directly interact with the Cys288 residue of Keap1, which partly accounts for its ability to induce Nrf2 activation and upregulate defensive gene expression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Compostos Alílicos/farmacologia , Cisteína/química , Proteínas do Citoesqueleto/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Sulfetos/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Western Blotting , Linhagem Celular , Células Cultivadas , Proteínas do Citoesqueleto/genética , Feminino , Humanos , Imuno-Histoquímica , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos , Fator 2 Relacionado a NF-E2/genética
18.
Amino Acids ; 46(1): 89-100, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23933994

RESUMO

Taurine is one of the most abundant non-essential amino acid in mammals and has many physiological functions in the nervous, cardiovascular, renal, endocrine, and immune systems. Upon inflammation, taurine undergoes halogenation in phagocytes and is converted to taurine chloramine (TauCl) and taurine bromamine. In the activated neutrophils, TauCl is produced by reaction with hypochlorite (HOCl) generated by the halide-dependent myeloperoxidase system. TauCl is released from activated neutrophils following their apoptosis and inhibits the production of inflammatory mediators such as, superoxide anion, nitric oxide, tumor necrosis factor-α, interleukins, and prostaglandins in inflammatory cells at inflammatory tissues. Furthermore, TauCl increases the expressions of antioxidant proteins, such as heme oxygenase 1, peroxiredoxin, thioredoxin, glutathione peroxidase, and catalase in macrophages. Thus, a central role of TauCl produced by activated neutrophils is to trigger the resolution of inflammation and protect macrophages and surrounding tissues from being damaged by cytotoxic reactive oxygen metabolites overproduced during inflammation. This is achieved by attenuating further production of proinflammatory cytokines and reactive oxygen metabolites and also by increasing the levels of antioxidant proteins that are able to scavenge and diminish the production of cytotoxic oxygen metabolites. These findings suggest that TauCl released from activated neutrophils may be involved in the recovery processes of cells affected by inflammatory oxidative stresses and thus TauCl could be used as a potential physiological agent to control pathogenic symptoms of chronic inflammatory diseases.


Assuntos
Citoproteção , Inflamação/metabolismo , Taurina/análogos & derivados , Animais , Antioxidantes/metabolismo , Apoptose , Doença Crônica , Humanos , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Macrófagos/patologia , Ativação de Neutrófilo , Neutrófilos/metabolismo , Neutrófilos/patologia , Oxirredutases/metabolismo , Taurina/metabolismo
19.
J Cell Sci ; 126(Pt 17): 4037-47, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23788426

RESUMO

Phagocytosis of apoptotic neutrophils, termed efferocytosis, is essential for the resolution of inflammation as it prevents the tissues surrounding the inflamed site from being exposed to the toxic contents of lytic cells. Resolvin D1 (RvD1), endogenously generated from docosahexaenoic acid during resolution of inflammation, is known to stimulate efferocytosis. However, the molecular mechanism underlying RvD1-mediated enhancement of efferocytosis remains largely unresolved. In the present study, murine macrophage-like RAW264.7 cells treated with lipopolysaccharide (LPS) exhibited markedly reduced efferocytic activity, but this was restored by co-incubation with RvD1. RvD1-induced restoration of the efferocytic activity appears to be mediated by downregulation of LPS-induced TNF-α expression. The inhibitory effect of RvD1 on LPS-induced TNF-α expression was associated with enhanced nuclear localization of p50/p50 homodimer and concomitant reduction of p65/p50 heterodimer accumulation in the nucleus. RvD1 triggered phosphorylation and proteasomal degradation of nuclear factor κB1 (NF-κB1) p105 to generate p50, which was subsequently translocated to the nucleus as a p50/p50 homodimer. Knockdown of NF-κB p50 abolished the ability of RvD1 to suppress TNF-α expression and also to restore efferocytosis, suggesting that the replacement of p65/p50 with p50/p50 homodimer in the nucleus is crucial for RvD1-mediated stimulation of efferocytosis. In a murine peritonitis model, intraperitoneal administration of RvD1 abolished the zymosan-A-induced TNF-α production, thereby stimulating efferocytosis. Taken together, these findings indicate that RvD1 expedites resolution of inflammation through induction of efferocytosis by p50/p50-homodimer-mediated repression of TNF-α production.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Fagocitose/imunologia , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/imunologia , Linhagem Celular , Regulação para Baixo , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Células Jurkat , Contagem de Leucócitos , Lipopolissacarídeos , Macrófagos , Camundongos , Camundongos Endogâmicos ICR , Neutrófilos/imunologia , Peritonite/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Zimosan
20.
Arch Pharm Res ; 36(6): 775-82, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23543651

RESUMO

Urethane, which is used as an anesthetic for animal experiments, causes inflammation and cancer in the lung. BALB/c mice received 1 mg/g of urethane once a week for four consecutive weeks via intraperitoneal injections developed interstitial infiltration of inflammatory cells and tumors in the lung. However, the intracellular signaling events which urethane causes inflammation and cancer are largely unknown. Here we show that urethane caused overproduction of reactive oxygen species (ROS) in RAW 264.7 macrophages and A549 lung epithelial cells. Pretreatment of these cells with the antioxidant N-acetylcysteine attenuated the urethane-induced ROS production. Urethane increased heme oxygenase-1 expression to protect these cells from cytotoxicity caused by overproduced ROS. In addition, urethane activated extracellular signal-regulated kinase (ERK) in both cell types. Overall, our data imply that urethane stimulates ROS production and ERK activation in macrophages and lung epithelial cells, and the overproduced ROS and activated ERK may promote tumor formation in the lung.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Pulmão/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Uretana/toxicidade , Acetilcisteína/farmacologia , Anestésicos Intravenosos/toxicidade , Animais , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Heme Oxigenase-1/metabolismo , Humanos , Pulmão/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...