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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 510(1): 156-162, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30678810

RESUMO

Locally applied radiation to the tumor is reported to stimulate systemic immune response. During radiotherapy to the abdominal cancer, spleen often receives certain dose, though as an important immune organ, little is known about the impact of splenic irradiation (SI) on systemic immune and local tumor control. Through a mice model, we found that the combination of SI with tumor irradiation (TI) helped in local control. The analysis of the tumor infiltrating leucocytes demonstrated that SI plus TI brought more T cell aggregation in the tumor microenvironment (TME), which helped in tumor control. Increased T cell infiltration may be partly due to higher expression of T cell chemokine in the TME and more expression of CXCR3 on the T cells in the spleen after SI. SI produced more IL-1ß in the spleen, IL-1ß stimulated the expression of CXCR3 on the T cells, and enhanced their migration ability. Taken together, radiation to the spleen combined with TI helped in local control through promoting T cell infiltration, and may be a considerable means to enhance the immunomodulatory of radiotherapy.


Assuntos
Neoplasias/radioterapia , Baço/efeitos da radiação , Animais , Movimento Celular/imunologia , Quimiocinas/metabolismo , Modelos Animais de Doenças , Interleucina-1beta/metabolismo , Linfócitos do Interstício Tumoral/citologia , Linfócitos do Interstício Tumoral/fisiologia , Camundongos , Neoplasias/prevenção & controle , Receptores CXCR3/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Resultado do Tratamento , Microambiente Tumoral/imunologia
2.
J Immunol ; 191(1): 500-8, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23733881

RESUMO

Critical roles of IL-27 in autoimmune diseases and infections have been reported; however, the contribution of endogenous IL-27 to tumor progression remains elusive. In this study, by using IL-27p28 conditional knockout mice, we demonstrate that IL-27 is critical in protective immune response against methyl-cholanthrene-induced fibrosarcoma and transplanted B16 melanoma, and dendritic cells (DCs) are the primary source. DC-derived IL-27 is required for shaping tumor microenvironment by inducing CXCL-10 expression in myeloid-derived suppressor cells and regulating IL-12 production from DCs, which lead to the recruitment and activation of NK and NKT cells resulting in immunological control of tumors. Indeed, reconstitution of IL-27 or CXCL-10 in tumor site significantly inhibits tumor growth and restores the number and activation of NK and NKT cells. In summary, our study identifies a previous unknown critical role of DC-derived IL-27 in NK and NKT cell-dependent antitumor immunity through shaping tumor microenvironment, and sheds light on developing novel therapeutic approaches based on IL-27.


Assuntos
Células Dendríticas/imunologia , Interleucinas/fisiologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/prevenção & controle , Células T Matadoras Naturais/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Células Dendríticas/metabolismo , Tolerância Imunológica , Células Matadoras Naturais/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Células T Matadoras Naturais/metabolismo
3.
Oncol Rep ; 51(3)2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38624021

RESUMO

It has been reported that PL2L60 proteins, a product of PIWIL2 gene which might be activated by an intragenic promoter, could mediate a common pathway specifically for tumorigenesis. In the present study, it was further identified by using western blot assay that the PL2L60 proteins could be degraded in cancer cells through a mechanism of selective autophagy in response to oxidative stress. The PL2L60 was downregulated in various types of cancer cells under the hypoxic condition independently of HIF­1α, resulting in apoptosis of cancer cells. Inhibition of autophagy by small interfering RNA targeting of either Beclin­1 (BECN1) or Atg5 resulted in restoration of PL2L60 expression in hypoxic cancer cell. The hypoxic degradation of PL2L60 was also blocked by the attenuation of the autophagosome membrane protein Atg8/microtubule­associated protein 1 light chain 3 (LC3) or autophagy cargo protein p62 expression. Surprisingly, Immunofluorescence analysis demonstrated that LC3 could be directly bound to PL2L60 and was required for the transport of PL2L60 from the nucleus to the cytoplasm for lysosomal flux under basal or activated autophagy in cancer cells. Moreover, flow cytometric analysis displayed that knocking down of PL2L60 mRNA but not PIWIL2 mRNA effectively inhibited cancer cell proliferation and promoted apoptosis of cancer cells. The similar results were obtained from in vivo tumorigenic experiment, in which PL2L60 downregulation in necroptosis areas was confirmed by immunohistochemistry. These results suggested that various cancer could be suppressed by promoting autophagy. The present study revealed a key role of autophagic degradation of PL2L60 in hypoxia­induced cancer cell death, which could be used as a novel therapeutic target of cancer.


Assuntos
Neoplasias , Humanos , RNA Interferente Pequeno/metabolismo , Hipóxia/metabolismo , Apoptose , Autofagia , Estresse Fisiológico , RNA Mensageiro , Proteínas Argonautas/metabolismo
4.
Am J Cancer Res ; 12(1): 265-279, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35141017

RESUMO

There are numerous antibodies used for cancer therapy in clinic, but they are essentially less efficacy than expected. None of them has tumor-specific and broad-spectral properties. PIWIL2-like (PL2L) protein 60 (PL2L60) is a product of alienated activation of PIWIL2 gene, and has been found to be specifically and widely expressed in various types of cancers, including hematopoietic and solid ones. Current study aims to investigate whether a monoclonal antibody (mAb) to PL2L60 has both tumor-specific and broad-spectral properties, which can be used universally to treat various types of cancers. The expression of PL2L60 protein in the cell surface and cytoplasm were determined in a panel of human and mouse tumor cell lines by flow cytometry, immunofluorescent microscopy and Western Blotting. The apoptosis and the cell cycle arrest of the tumor cells treated with mAb KAO3 were evaluated by flow cytometry. The tumorigenesis of the mAb KAO3-pretreated tumor cells was determined by tumor incidence and tumor size, and the efficacy of mAb KAO3 treatment on tumor growth in tumors-bearing mice were kinetically evaluated. Complement-dependent cytotoxicity (CDC) assay was used to determine the capacity of mAb KAO3 to kill tumor cells. Treatment of human or mouse tumor cells from hematopoietic or solid tumors with mAb KAO3 at the time of inoculation efficiently inhibited tumorigenesis in the severe combined immunodeficient (SCID) mice. Moreover, injection of mAb KAO3 into established tumors significantly inhibited their growth, and prolonged survival of the tumor-bearing mice, including lymphoma, breast cancer, lung cancer and cervical cancer. The efficacy of mAb KAO3 treatment is likely associated with its binding to PL2L60 expressed on tumor cell surface, which may lead to cancer cell death through blocking cell cycling and/or activation of complement. In conclusion, we have identified a tumor-specific mAb to PL2L60 (KAO3), which may be used potentially to treat all the types of human cancers including from both hematopoietic and solid ones.

5.
J Exp Med ; 202(8): 1141-51, 2005 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-16230479

RESUMO

The Scurfy mutation of the FoxP3 gene (FoxP3(sf)) in the mouse and analogous mutations in human result in lethal autoimmunity. The mutation of FoxP3 in the hematopoietic cells impairs the development of regulatory T cells. In addition, development of the Scurfy disease also may require mutation of the gene in nonhematopoietic cells. The T cell-extrinsic function of FoxP3 has not been characterized. Here we show that the FoxP3(sf) mutation leads to defective thymopoiesis, which is caused by inactivation of FoxP3 in the thymic stromal cells. FoxP3 mutation also results in overexpression of ErbB2 in the thymic stroma, which may be involved in defective thymopoiesis. Our data reveal a novel T cell-extrinsic function of FoxP3. In combination, the T cell-intrinsic and -extrinsic defects provide plausible explanation for the severity of the autoimmune diseases in the scurfy mice and in patients who have immunodysregulation, polyendocrinopathy, enteropathy, and X-linked syndrome.


Assuntos
Doenças Autoimunes/genética , Fatores de Transcrição Forkhead/genética , Doenças Linfáticas/genética , Mutação/genética , Timo/crescimento & desenvolvimento , Timo/patologia , Animais , Apoptose/fisiologia , Doenças Autoimunes/patologia , Bromodesoxiuridina , Primers do DNA , Citometria de Fluxo , Luciferases , Doenças Linfáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/metabolismo , Timo/citologia , Timo/metabolismo
6.
J Exp Med ; 195(8): 959-71, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11956287

RESUMO

A number of in vitro studies have suggested that costimulatory molecules B7-1 and B7-2 and their receptor CD28 can promote clonal deletion, and limited in vivo studies have indicated that CD28 is involved in the clonal deletion of some T cells. However, the significance of B7-mediated clonal deletion in preventing autoimmune diseases has not been studied systematically. Here we report that the perinatal blockade of B7-1 and B7-2 substantially inhibits the clonal deletion of T cells in the thymus and leads to an accumulation of T cells capable of inducing fatal multiorgan inflammation. These results reveal a critical role for costimulatory molecules B7-1 and B7-2 in deleting pathogenic autoreactive T cells in the thymus. The critical role of B7-1 and B7-2 in T cell clonal deletion may explain, at least in part, the paradoxical increase of autoimmune disease in mice deficient for this family of costimulatory molecules, such as cytotoxic T lymphocyte associated molecule 4, CD28, and B7-2. The strong pathogenicity of the self-reactive T cells supports a central hypothesis in immunology, which is that clonal deletion plays an important role in preventing autoimmune diseases.


Assuntos
Antígenos CD/imunologia , Antígeno B7-1/imunologia , Deleção Clonal/imunologia , Glicoproteínas de Membrana/imunologia , Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos Virais/imunologia , Antígeno B7-2 , Contagem de Linfócito CD4 , Relação CD4-CD8 , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/biossíntese , Feminino , Intestinos/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Pâncreas/imunologia
7.
J Exp Med ; 197(12): 1721-30, 2003 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-12810690

RESUMO

B7H1 (PDL1) and B7DC (PDL2) are two new members of the B7 family that can interact with PD-1, a putative negative regulator for immune function. Recent studies have provided evidence for inhibitory functions of both members via PD-1. Meanwhile, compelling evidence exists for costimulatory function of both members. Here we demonstrate that expression of B7DC on the tumor cells promotes CD8 T cell-mediated rejection of tumor cells, at both the induction and effector phase of antitumor immunity. Moreover, B7DC binds to PD-1(-/-) cells and enhances T cell killing in a PD-1-independent mechanism. Our results demonstrate a novel pathway for B7DC to promote tumor immunity and may reconcile the apparently contradictory findings on the function of B7DC.


Assuntos
Antígenos de Superfície , Antígeno B7-1/imunologia , Proteínas Sanguíneas , Linfócitos T CD8-Positivos/imunologia , Neoplasias Experimentais/imunologia , Peptídeos , Proteínas/imunologia , Transferência Adotiva , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Proteínas Reguladoras de Apoptose , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígeno B7-H1 , Linfócitos T CD8-Positivos/metabolismo , Células CHO , Cricetinae , Citotoxicidade Imunológica , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Transplante de Neoplasias , Proteína 2 Ligante de Morte Celular Programada 1 , Receptor de Morte Celular Programada 1 , Proteínas/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Baço/citologia , Baço/imunologia , Linfócitos T Citotóxicos/imunologia , Transgenes , Quimeras de Transplante , Células Tumorais Cultivadas
8.
Biochem J ; 416(2): 179-87, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18588516

RESUMO

Two of the most common signalling pathways in breast cancer are the ER (oestrogen receptor) ligand activation pathway and the E-cadherin snai1 slug EMT (epithelial-mesenchymal transition) pathway. Although these pathways have been thought to interact indirectly, the present study is the first to observe direct interactions between these pathways that involves the regulation of slug expression. Specifically we report that ligand-activated ERalpha suppressed slug expression directly by repression of transcription and that knockdown of ERalpha with RNA interference increased slug expression. More specifically, slug expression was down-regulated in ERalpha-negative MDA-MB-468 cells transfected with ERalpha after treatment with E2 (17beta-oestradiol). The down-regulation of slug in the ERalpha-positive MCF-7 cell line was mediated by direct repression of slug transcription by the formation of a co-repressor complex involving ligand-activated ERalpha protein, HDAC1 (histone deacetylase 1) and N-CoR (nuclear receptor co-repressor). This finding was confirmed by sequential ChIP (chromatin immunoprecipitation) studies. In the MCF-7 cell line, slug expression normally was low. In addition, knockdown of ERalpha with RNA interference in this cell line increased slug expression. This effect could be partially reversed by treatment of the cells with E2. The efficacy of the effect of ERalpha on slug repression was dependent on the overall level of ERalpha. These observations confirmed that slug was an E2-responsive gene.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Fatores de Transcrição/genética , Transcrição Gênica , Sequência de Bases , Neoplasias da Mama , Linhagem Celular Tumoral , Primers do DNA , DNA Complementar/genética , Receptor alfa de Estrogênio/deficiência , Feminino , Humanos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Neoplásico/genética , RNA Neoplásico/isolamento & purificação , Fatores de Transcrição da Família Snail , Transfecção
9.
J Gastroenterol Hepatol ; 23(7 Pt 2): e231-5, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17683476

RESUMO

BACKGROUND AND AIM: Both nuclear factor kappa B (NF-kappaB) activation and inducible nitric oxide synthase (iNOS) expression increase in the liver injury, and there are NF-kappaB binding sites in the iNOS promoter. The aim of this study was to investigate the correlation between iNOS expression and NF-kappaB activation in hepatitis induced by concanavalin A (con A). METHODS: Eighty-eight male BALB/c mice were randomly divided into three groups: vehicle control group, con A group and pyrrolidine dithiocarbamate (PDTC) plus con A group. In the vehicle control group, the mice were treated with saline (0.3 mL, i.v.). In the con A group, the mice were treated with con A (20 mg/kg, i.v.). In the PDTC + con A group, the mice were pretreated with PDTC (120 mg/kg, i.p.) 30 min before administration of con A (20 mg/kg, i.v.). Blood samples were taken from the retro-orbital venous plexus at 0.5, 1, 4, 8 and 16 h after con A injection and the mice were killed immediately. The plasma alanine aminotransferase (ALT) levels were measured by the standard photometric method. Nitric oxide (NO) levels in the liver homogenate were assayed by spectroscopy. Liver tissues were sectioned and stained with hematoxylin-eosin for histological examination. Activation of NF-kappaB, degradation of inhibitor of kappa B alpha (IkappaBalpha), and expression of iNOS were measured by western blot. RESULTS: In the con A group, the plasma ALT activity and NO levels in the liver increased significantly at 1 h (P < 0.05, n = 8) and reached a peak at 4 h after con A injection. The liver injury in this group was characterized by liver necrosis, cell swelling and fatty degeneration. Cytosolic IkappaBalpha decreased slightly at 30 min after con A challenge, was undetectable at 1 h and reappeared at 4 h. Correspondingly, the NF-kappaB level in the nucleus was highest at 1 h. The iNOS expression increased at 30 min after con A injection and reached a maximum at 4 h. Pretreatment with PDTC prevented these changes and attenuated the liver injury. CONCLUSION: Con A-induced iNOS expression in the liver is dependent on the activation of NF-kappaB.


Assuntos
Hepatite/enzimologia , Fígado/enzimologia , Óxido Nítrico Sintase Tipo II/biossíntese , Fator de Transcrição RelA/metabolismo , Transporte Ativo do Núcleo Celular , Alanina Transaminase/sangue , Animais , Concanavalina A , Modelos Animais de Doenças , Indução Enzimática , Hepatite/etiologia , Hepatite/patologia , Proteínas I-kappa B/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Inibidor de NF-kappaB alfa , Necrose , Óxido Nítrico/metabolismo , Pirrolidinas/farmacologia , Índice de Gravidade de Doença , Tiocarbamatos/farmacologia , Fatores de Tempo , Fator de Transcrição RelA/antagonistas & inibidores , Regulação para Cima
10.
J Leukoc Biol ; 81(6): 1422-33, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17332372

RESUMO

NK dendritic cells (DCs; NKDCs) appear to emerge as a distinct DC subset in humans and rodents, which have the functions of NK cells and DCs. However, the developmental relationship of NKDCs (CD11c(+)NK1.1(+)) to CD11c(+)NK1.1(-) DCs has not been addressed. Herein, we show that NKDCs exist exclusively in the compartment of CD11c(+)MHC II(-) cells in the steady state and express variable levels of DC subset markers, such as the IFN-producing killer DC marker B220, in a tissue-dependent manner. They can differentiate into NK1.1(-) DCs, which is accompanied by the up-regulation of MHC Class II molecules and down-regulation of NK1.1 upon adoptive transfer. However, NK cells (NK(+)CD11c(-)) did not differentiate into NK1.1(+)CD11c(+) cells upon adoptive transfer. Bone marrow-derived Ly6C(+) monocytes can be a potential progenitor of NKDCs, as some of them can differentiate into CD11c(+)NK1.1(+) as well as CD11c(+)NK1.1(-) cells in vivo. The steady-state NKDCs have a great capacity to lyse tumor cells but little capability to present antigens. Our studies suggest that NKDCs are an intermediate of developing DCs. These cells appear to bear the unique surface phenotype of CD11c(+)NK1.1(+)MHC II(-) and possess strong cytotoxic function yet show a poor ability to present antigen in the steady state. These findings suggest that NKDCs may play a critical role in linking innate and adaptive immunity.


Assuntos
Antígeno CD11c/imunologia , Células Dendríticas/citologia , Células Dendríticas/fisiologia , Células Matadoras Naturais/citologia , Transferência Adotiva , Animais , Apresentação de Antígeno , Diferenciação Celular , Linhagem Celular , Linhagem Celular Tumoral , Linhagem da Célula , Proliferação de Células , Sobrevivência Celular , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Feminino , Antígenos de Histocompatibilidade Classe II/imunologia , Imunidade Ativa , Imunidade Inata , Células Matadoras Naturais/imunologia , Antígenos Comuns de Leucócito/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/imunologia
11.
Sci Rep ; 8(1): 11934, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-30093664

RESUMO

Endogenous immune response participates in tumor control, and radiotherapy has immune modulatory capacity, but the role of immune modulation in the tumor microenvironment invoked by radiotherapy in radiosensitivity is poorly defined. In the present study, a radio-resistant melanoma cell line was obtained after repeated irradiation to the parental tumor in C57BL/6 mice. Radiotherapy resulted in aggregation of CD8+ and CD3+ T cells, and decrease of myeloid-derived suppressor cells and dendritic cells in the parental tumor, but not in the resistant tumors. CD4+ T cells and B cells did not change significantly. The CD8+ T cell infiltration after radiotherapy is important for tumor response, because in the nude mice and CD8+ T cell-depleted C57BL/6 mice, the parental and resistant tumor has similar radiosensitivity. Patients with good radiation response had more CD8+ T cells aggregation after radiotherapy. Radiotherapy resulted in robust transcription of T cell chemoattractant in the parental cells, and the expression of CCL5 was much higher. These results reveal a novel mechanism of radioresistance, tumor cells inhibit the infiltration of CD8+ T cell after radiotherapy and become radioresistant. Increasing CD8+ T cell infiltration after RT may be an effective way to improve tumor radiosensitivity.


Assuntos
Linfócitos T CD8-Positivos/efeitos da radiação , Melanoma Experimental/radioterapia , Tolerância a Radiação/efeitos da radiação , Microambiente Tumoral/efeitos da radiação , Animais , Apoptose/genética , Apoptose/imunologia , Apoptose/efeitos da radiação , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Sobrevivência Celular/efeitos da radiação , Quimiocinas/genética , Quimiocinas/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Camundongos Endogâmicos C57BL , Camundongos Nus , Tolerância a Radiação/imunologia , Carga Tumoral/genética , Carga Tumoral/imunologia , Carga Tumoral/efeitos da radiação , Microambiente Tumoral/imunologia
12.
Front Microbiol ; 8: 1839, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29085341

RESUMO

Cronobacter sakazakii is an important foodborne pathogen that causes neonatal meningitis and sepsis, with high mortality in neonates. However, very little information is available regarding the pathogenesis of C. sakazakii at the genetic level. In our previous study, a cellulose biosynthesis-related gene (bcsR) was shown to be involved in C. sakazakii adhesion/invasion into epithelial cells. In this study, the detailed functions of this gene were investigated using a gene knockout technique. A bcsR knockout mutant (ΔbcsR) of C. sakazakii ATCC BAA-894 showed decreased adhesion/invasion (3.9-fold) in human epithelial cell line HCT-8. Biofilm formation by the mutant was reduced to 50% of that exhibited by the wild-type (WT) strain. Raman spectrometry was used to detect variations in biofilm components caused by bcsR knockout, and certain components, including carotenoids, fatty acids, and amides, were significantly reduced. However, another biofilm component, cellulose, was increased in ΔbcsR, suggesting that bcsR negatively affects cellulose biosynthesis. This result was also verified via RT-PCR, which demonstrated up-regulation of five crucial cellulose synthesis genes (bcsA, B, C, E, Q) in ΔbcsR. Furthermore, the expression of other virulence or biofilm-related genes, including flagellar assembly genes (fliA, C, D) and toxicity-related genes (ompA, ompX, hfq), was studied. The expression of fliC and ompA in the ΔbcsR mutant was found to be remarkably reduced compared with that in the wild-type and the others were also affected excepted ompX. In summary, bcsR is a negative regulator of cellulose biosynthesis but positively regulates biofilm formation and the adhesion/invasion ability of C. sakazakii.

13.
Oncotarget ; 8(28): 46104-46120, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-28545024

RESUMO

PIWIL2-like (PL2L) protein 60 (PL2L60), a product of aberrantly activated PIWIL2 gene, is widely expressed in various types of tumors and may promote tumorigenesis. However, the mechanisms underlying the activation of expression of PL2L60 remain unknown. In this study, an intragenic promoter responsible for the activation of PL2L60 within the human PIWIL2 gene has been identified, cloned and characterized. The promoter of PL2L60 is located in the intron 10 of the host gene PIWIL2. Bioinformatic and mutagenic analysis reveals that this intragenic promoter within the sequence of 50 nucleotides contains two closely arranged cis-acting elements specific for the hepatic leukemia factor (HLF) in the positive strand and signal transducer and activator of transcription 3 (STAT3) in the negative strand. Chromatin immunoprecipitation analysis demonstrates that both the HLF and polymerase II (Pol II), a hallmark of active promoters, directly bind to the sequence, although STAT3 does not. Knockdown of HLF and STAT3 alone or both by RNA interference significantly reduced both promoter activity and the PL2L60 protein expression, although there is no additive effect. The expression of PL2L60 proteins was enhanced when host gene Piwil2 was genetically disrupted in a murine cell model. Taken together, we have identified a PL2L60-specific intragenic promoter in the host gene of PIWIL2, which is interdependently activated by HLF and STAT3 through steric interaction. This activation is dependent on cellular milieu rather than the integrity of host gene PIWIL2, highlighting a novel, important mechanism for a cancer-causing gene to be activated during tumorigenesis.


Assuntos
Proteínas Argonautas/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Carcinogênese/genética , Regulação Neoplásica da Expressão Gênica , Oncogenes/genética , Regiões Promotoras Genéticas/genética , Fator de Transcrição STAT3/metabolismo , Regulação Alostérica , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/genética , Sequências Reguladoras de Ácido Nucleico/genética , Fator de Transcrição STAT3/genética
14.
Theranostics ; 6(8): 1232-43, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27279914

RESUMO

Both adaptive and acquired resistance significantly limits the efficacy of the epidermal growth factor receptor (EGFR) kinase inhibitors. However, the distinct or common mechanisms of adaptive and acquired resistance have not been fully characterized. Here, through systematic modeling of erlotinib resistance in lung cancer, we found that feedback reactivation of MAPK signaling following erlotinib treatment, which was dependent on the MET receptor, contributed to the adaptive resistance of EGFR inhibitors. Interestingly, acquired resistance to erlotinib was also associated with the MAPK pathway activation as a result of CRAF or NRAS amplification. Consequently, combined inhibition of EGFR and MAPK impeded the development of both adaptive and acquired resistance. These observations demonstrate that adaptive and acquired resistance to EGFR inhibitors can converge on the same pathway and credential cotargeting EGFR and MAPK as a promising therapeutic approach in EGFR mutant tumors.


Assuntos
Antineoplásicos/farmacologia , Resistência a Medicamentos , Receptores ErbB/antagonistas & inibidores , Cloridrato de Erlotinib/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Animais , Carcinoma Pulmonar de Células não Pequenas , Linhagem Celular Tumoral , Receptores ErbB/genética , Xenoenxertos , Humanos , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno/genética
15.
Int J Oncol ; 48(6): 2461-71, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27035405

RESUMO

Protein arginine methylation is a common posttranslational modification resulting in the generation of asymmetric dimethylarginine (aDMA) and symmetric dimethylarginine (sDMA). Currently, the regulation of aDMA or sDMA by hypoxia, nutrient stavation or cytokines in the tumor microenvironment remains largely unknown. Here we show that p90aDMA, p70aDMA and p90sDMA, endogenous proteins containing aDMA or sDMA with mass 70 or 90 kDa, were widely and dominantly expressed in breast cancer cell lines. Notably, it was p90aDMA rather than p90sDMA that accumulated in the nucleus upon stimulation of cancer cells with interleukin (IL)-2, IL-4, IL-6 but not IL-8. In addition, the p90aDMA accumulation could be inhibited after treatment with a global methyltrasferase inhibitor, adenosine-2',3'-dialdehyde (AdOx). It seemed that some endogenous proteins in cancer cells were asymmetrically arginine-methylated upon exposure to some cytokines.. Furthermore, endogenous proteins of aDMA, such as p90aDMA and p70aDMA, were degraded in response to hypoxia, nutrient starvation and rapamycin treatment in breast and cervical cancer cells. IL-2/4/6 slightly increased basal autophagy but slightly decreased the rapamycin­induced autophagy in cancer cells, suggesting that IL-2/4/6 and autophagy inducers play distinct roles in the regulation of aDMA of proteins. Conversely, rapamycin accumulated p90sDMA in MDA-MB­231 and MCF-7 cells. Taken together, our results add a new dimension to the complexity of arginine methylated regulation in response to various stimuli and provide the first evidence that aDMA serves as one specific degradation signal of selective autophagy.


Assuntos
Arginina/análogos & derivados , Neoplasias da Mama/metabolismo , Interleucina-2/farmacologia , Interleucina-4/farmacologia , Interleucina-6/farmacologia , Proteínas Nucleares/metabolismo , Arginina/metabolismo , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Células MCF-7 , Peso Molecular , Proteínas Nucleares/química , Processamento de Proteína Pós-Traducional , Sirolimo/farmacologia , Microambiente Tumoral , Neoplasias do Colo do Útero/metabolismo
16.
Sci Rep ; 6: 20489, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26841847

RESUMO

The activation/inactivation of HIF1α is precisely regulated in an oxygen-dependent manner. HIF1α is essential for hypoxia induced apoptosis and cell cycle arrest. Several recent studies indicated that the expression of miRNAs can be modulated by hypoxia. However, the involvement of miRNAs in the regulation of HIF1α induction remains elusive. In present study, we demonstrated that miR-101 was rapidly and transiently induced after hypoxia in breast cancer cells. Over-expression of miR-101 significantly inhibited cell proliferation in breast cancer cells through increased apoptosis and cell cycle arrest in normoxia condition. This inhibitory phenomenon seems due to miR-101-mediated induction of HIF1α, because we identified that VHL, a negative regulator of HIF1α, is a novel target of miR-101 and over-expression of miR-101 decreased VHL levels and subsequently stabilized HIF1α and induced its downstream target VEGFA. Furthermore, we demonstrated that siRNA-mediated knockdown of VHL or HIF1α overexpression could also induce apoptosis and cell cycle arrest whereas enforced expression of VHL, administration of anti-miR-101 oligos or treatment of 2-MeOE2, an inhibitor of HIF1α, could rescue cells from such inhibition. These results reveal a novel regulatory mechanism of HIF1α induction in normoxia and suggest that miR-101 mediated proliferation inhibition may through HIF1α mediated apoptosis and cell cycle arrest.


Assuntos
Neoplasias da Mama/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , MicroRNAs/genética , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Regiões 3' não Traduzidas , Apoptose , Neoplasias da Mama/metabolismo , Pontos de Checagem do Ciclo Celular , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Hipóxia Tumoral , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
17.
Sci Rep ; 6: 24728, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27090737

RESUMO

Abnormally high activation of transforming growth factor-ß (TGF-ß) signaling has been demonstrated to be involved in the initiation and progression of keloids. However, the functional role of long non-coding RNA (lncRNA)-activated by TGF-ß (lncRNA-ATB) in keloids has not been documented. Here we investigated the role of lncRNA-ATB in the autocrine secretion of TGF-ß in keloid fibroblasts (KFs) and explored the underlying molecular mechanism. Using immunohistochemistry and quantitative RT-PCR analysis, we showed that lncRNA-ATB and ZNF217, a transcriptional activator of TGF-ß, were overexpressed and miR-200c, which targets ZNF217, was under-expressed in keloid tissue and keloid fibroblasts. Through gain- and loss-of-function studies, we demonstrated that knockdown of lncRNA-ATB decreased autocrine secretion of TGF-ß2 and ZNF217 expression but upregulated expression of miR-200c in KFs. Stable downregulation of ZNF217 expression decreased the autocrine secretion of TGF-ß2. miR-200c was endogenously associated with lncRNA-ATB, and inhibition of miR-200c overcame the decrease in ZNF217 expression in KFs. Taken together, these findings indicate that lncRNA-ATB governs the autocrine secretion of TGF-ß2 in KFs, at least in part, by downregulating the expression level of ZNF217 via miR-200c, suggesting a signaling axis consisting of lncRNA-ATB/miR-200c/ZNF217/TGF-ß2. These findings may provide potential biomarkers and targets for novel diagnostic and therapeutic approaches for keloids.


Assuntos
Queloide/metabolismo , MicroRNAs/genética , RNA Longo não Codificante/genética , Transativadores/genética , Fator de Crescimento Transformador beta2/metabolismo , Biomarcadores , Regulação para Baixo , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Humanos
18.
Sci Rep ; 6: 32199, 2016 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-27599451

RESUMO

Acute kidney injury (AKI) is a common complication after severe burns. Melatonin has been reported to protect against multiple organ injuries by increasing the expression of SIRT1, a silent information regulator that regulates stress responses, inflammation, cellular senescence and apoptosis. This study aimed to investigate the protective effects of melatonin on renal tissues of burned rats and the role of SIRT1 involving the effects. Rat severely burned model was established, with or without the administration of melatonin and SIRT1 inhibitor. The renal function and histological manifestations were determined to evaluate the severity of kidney injury. The levels of acetylated-p53 (Ac-p53), acetylated-p65 (Ac-p65), NF-κB, acetylated-forkhead box O1 (Ac-FoxO1), Bcl-2 and Bax were analyzed to study the underlying mechanisms. Our results suggested that severe burns could induce acute kidney injury, which could be partially reversed by melatonin. Melatonin attenuated oxidative stress, inflammation and apoptosis accompanied by the increased expression of SIRT1. The protective effects of melatonin were abrogated by the inhibition of SIRT1. In conclusion, we demonstrate that melatonin improves severe burn-induced AKI via the activation of SIRT1 signaling.


Assuntos
Injúria Renal Aguda/prevenção & controle , Queimaduras/complicações , Melatonina/farmacologia , Sirtuína 1/metabolismo , Acetilação/efeitos dos fármacos , Injúria Renal Aguda/etiologia , Animais , Apoptose/efeitos dos fármacos , Citocinas/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Ratos Sprague-Dawley , Proteína Supressora de Tumor p53/metabolismo
19.
Sheng Li Xue Bao ; 57(6): 755-60, 2005 Dec 25.
Artigo em Zh | MEDLINE | ID: mdl-16344902

RESUMO

Roscovitine is a specific inhibitor of cyclin-dependent kinases (cdks) cdc2/cyclin B, cdk2/cyclin A, cdk2/cyclin E and cdk5/p35. The studies on the enzyme inhibitory properties and cellular effects of roscovitine revealed that it arrests cells in G(2)/M and G(1)/S phase, inhibits the proliferation of mammalian cells and induces cell death. However, the characteristics of cell death and exact mechanism by which this cdk inhibitor kills transformed cells are unknown. We previously investigated that the roscovitine induces apoptotic death of mitotic PC12 cells. The present study was to identify whether the roscovitine-induced death is related with the specific elements of caspases in pathway of apoptosis. The morphological data of caspase-3 immunofluorocytochemistry double staining with hoechst 33342 indicated that apoptotic nuclei were identified as nuclei with chromatin condensation and nuclear fragmentation, and that caspase-3 active p17 subunit co-existed in PC12 cells treated with roscovitine 50 micromol/L for 4 h. The number of the caspase-3 positive cells increased significantly to about 42%, as compared with the normal control (P<0.001). The data of MTT assay showed that the number of viable cells treated by roscovitine (50 micromol/L) alone for 12 h was 29.03%, of the untreated controls. Both a broad-spectrum caspase inhibitor Z-VAD-FMK (50 mumol/L) and a specific caspase-3 inhibitor Z-DEVD-FMK (100 micromol/L) increased viable PC12 cells to 45.16%, (Z-DEVD-FMK) and 58.06%, (Z-VAD-FMK), respectively, in the presence of roscovitine. Non-erythroid a-spectrin is a cytoskeleted protein that is a substrate of caspase-3 cysteine proteases. To confirm the activity of caspase-3 that produced in roscovitine (50 micromol/L for 12 h)-induced PC12 cell death, activated caspase-3 specific 120 kDa spectrin breakdown products (SBDP) were detected by Western bloting using the mouse anti-non-erythroid a-spectrin monoclonal antibody. The mean relative density of bands corresponding to caspase-3 specific SBDP levels were significantly increased in the cytosolic fractions treated with roscovitine, as compared to the normal control (P<0.001). These results indicate that caspase signals, especially caspase-3 signal are necessary for the progression of proliferating PC12 cell apoptotic death evoked by roscovintine.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Animais , Apoptose/fisiologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Células PC12 , Ratos , Roscovitina
20.
Am J Cancer Res ; 5(9): 2626-42, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26609472

RESUMO

Autophagy has dual functions in cell survival and death. However, the effects of autophagy on cancer cell survival or death remain controversial. In this study, we show that Autophagy can mediate programmed cell death (PCD) of cancer cells in responding to cobalt chloride (CoCl2)-induced hypoxia in a Beclin-1-independent but autophagy protein 5 (ATG5)-dependent manner. Although ATG5 is not directly induced by CoCl2, its constitutive expression is essential for CoCl2-induced PCD. The ATG5-mediated autophagic PCD requires interplays with endoplasmic reticulum (ER) and/or mitochondria. In this process, ATG5 plays a central role in regulating ER stress protein CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) and mitochondrial protein second mitochondria derived activator of caspases (Smac). Two pathways for autophagic PCD in cancer cells responding to hypoxia have been identified: ATG5/CHOP/Smac pathway and ATG5/Smac pathway, which are probably dependent on the context of cell lines. The former is more potent than the latter for the induction of PCD at the early stage of hypoxia, although the ultimate efficiency of both pathways is comparable. In addition, both pathways may require ATG5-mediated conversion of LC3-I into LC3-II. Therefore, we have defined two autophagy-mediated pathways for the PCD of cancer cells in hypoxia, which are dependent on ATG5, interplayed with ER and mitochondria and tightly regulated by hypoxic status. The findings provide a new evidence that autophagy may inhibit tumor cell proliferation through trigger of PCD, facilitating the development of novel anti-cancer drugs.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA