Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Stress ; 3(11): 348-360, 2019 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-31799501

RESUMO

LTX-315 is an oncolytic peptide that has antitumor efficacy in mice grafted with various tumor cell lines and is currently being tested in phase II clinical trials. Here we aimed to further evaluate LTX-315 in conditional genetic mouse models of cancer that typically resist current treatment options and to better understand the drug's mode of action in vivo. We report LTX-315 mediates profound antitumor effects against Braf- and Pten-driven melanoma and delays the progression of Kras- and P53-driven soft tissue sarcoma in mice. Additionally, we show in melanoma that LTX-315 triggers two sequential phases of antitumor response. The first phase of response, which begins within minutes of drug delivery into tumors, is defined by disrupted tumor vasculature and decreased tumor burden and occurs independently of lymphocytes. The second phase of response, which continues over weeks, is defined by long-term alteration of the tumor microenvironment; the changes induced by LTX-315 are most notably characterized by CD8+ T cell infiltration. We further show that these CD8+ T cells are involved in suppressing melanoma outgrowth in mice and report similar CD8+ T cell infiltration following LTX-315 treatment in melanoma and sarcoma patients. Taken together, these findings reveal LTX-315's multiple antitumor effects, including disrupting the tumor vasculature and promoting the conversion of poorly immunogenic tumors into ones that display antitumor T cell immunity.

2.
Am J Cancer Res ; 9(1): 180-185, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30755821

RESUMO

Methylation at Arg198 and Arg200 residues of the EGFR extracellular domain by PRMT1 have been demonstrated to enhance EGFR activation by the canonical ligands, EGF and TGFα. On the other hand, RNase 5 was identified as a new ligand of EGFR recently. However, the interplay between EGFR methylation and RNase 5 in EGFR activation is still unclear. Here, we showed that RNase 5 activated EGFR and enhanced cell proliferation in colorectal cancer cells. PRMT1 positively regulated EGFR signaling activation by RNase 5. Inhibition of EGFR methylation by methylation-site mutagenesis reduced the binding affinity of RNase 5 to EGFR and abrogated RNase 5-mediated EGFR activation, suggesting that PRMT1-mediated EGFR methylation is critical for EGFR activation by RNase 5. Notably, RNase 5 diminished the inhibitory activity of cetuximab on colorectal cancer cells, implying RNase 5 is a potential biomarker to predict cetuximab response in colorectal cancer.

3.
Nat Commun ; 9(1): 1908, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29765039

RESUMO

Enriched PD-L1 expression in cancer stem-like cells (CSCs) contributes to CSC immune evasion. However, the mechanisms underlying PD-L1 enrichment in CSCs remain unclear. Here, we demonstrate that epithelial-mesenchymal transition (EMT) enriches PD-L1 in CSCs by the EMT/ß-catenin/STT3/PD-L1 signaling axis, in which EMT transcriptionally induces N-glycosyltransferase STT3 through ß-catenin, and subsequent STT3-dependent PD-L1 N-glycosylation stabilizes and upregulates PD-L1. The axis is also utilized by the general cancer cell population, but it has much more profound effect on CSCs as EMT induces more STT3 in CSCs than in non-CSCs. We further identify a non-canonical mesenchymal-epithelial transition (MET) activity of etoposide, which suppresses the EMT/ß-catenin/STT3/PD-L1 axis through TOP2B degradation-dependent nuclear ß-catenin reduction, leading to PD-L1 downregulation of CSCs and non-CSCs and sensitization of cancer cells to anti-Tim-3 therapy. Together, our results link MET to PD-L1 stabilization through glycosylation regulation and reveal it as a potential strategy to enhance cancer immunotherapy efficacy.


Assuntos
Antígeno B7-H1/imunologia , Hexosiltransferases/imunologia , Evasão da Resposta Imune , Proteínas de Membrana/imunologia , Neoplasias/imunologia , Células-Tronco Neoplásicas/imunologia , Animais , Antígeno B7-H1/genética , Linhagem Celular Tumoral , DNA Topoisomerases Tipo II/genética , DNA Topoisomerases Tipo II/imunologia , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Hexosiltransferases/genética , Humanos , Proteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neoplasias/genética , Neoplasias/fisiopatologia , Células-Tronco Neoplásicas/citologia , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas de Ligação a Poli-ADP-Ribose/imunologia , beta Catenina/genética , beta Catenina/imunologia
4.
Breast Cancer ; 25(1): 74-80, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28643125

RESUMO

BACKGROUND: Epidermal growth factor receptor (EGFR) is often overexpressed in triple-negative breast cancer (TNBC). However, clinical studies have shown that therapies against EGFR are not effective in patients with TNBC. Recently, it has been reported that arginine 198/200 in EGFR extracellular domain is methylated by PRMT1 and that the methylation confers resistance to EGFR monoclonal antibody cetuximab in colorectal cancer cells. To explore a potential mechanism underlying intrinsic resistance to anti-EGFR therapy in TNBC, we investigated the role of PRMT1 in EGFR methylation and signaling in MDA-MB-468 (468) TNBC cells. METHODS: We knocked down PRMT1 in 468 cells by shRNA, and subjected the cell lysates to Western blot analysis to examine EGFR activation and its downstream molecules. We also evaluated cell proliferation and sphere formation of PRMT1-knockdown cells. Finally, we examined the effects of pan-PRMT inhibitor, AMI-1, on cetuximab by colony formation and soft agar assays. RESULTS: EGFR methylation and activity was significantly reduced in PRMT1-knockdown cells compared to the parental cells. Knockdown of PRMT1 also reduced cell proliferation and sphere formation. Moreover, AMI-1 sensitized 468 cells to cetuximab. CONCLUSION: The results indicate that PRMT1 is critical for EGFR activity in 468 cells. Our data also suggest that inhibition of PRMT1 sensitizes TNBC cells to cetuximab. Thus, inhibition of PRMT1 may be an effective therapeutic strategy to overcome intrinsic resistance to cetuximab in TNBC.


Assuntos
Metilação de DNA , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células/efeitos dos fármacos , Cetuximab/farmacologia , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Prognóstico , Inibidores de Proteínas Quinases/farmacologia , Proteína-Arginina N-Metiltransferases/genética , Proteínas Repressoras/genética , Transdução de Sinais/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
5.
Science ; 358(6367)2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29191879

RESUMO

Bone marrow-derived myeloid cells can accumulate within tumors and foster cancer outgrowth. Local immune-neoplastic interactions have been intensively investigated, but the contribution of the systemic host environment to tumor growth remains poorly understood. Here, we show in mice and cancer patients (n = 70) that lung adenocarcinomas increase bone stromal activity in the absence of bone metastasis. Animal studies reveal that the cancer-induced bone phenotype involves bone-resident osteocalcin-expressing (Ocn+) osteoblastic cells. These cells promote cancer by remotely supplying a distinct subset of tumor-infiltrating SiglecFhigh neutrophils, which exhibit cancer-promoting properties. Experimentally reducing Ocn+ cell numbers suppresses the neutrophil response and lung tumor outgrowth. These observations posit osteoblasts as remote regulators of lung cancer and identify SiglecFhigh neutrophils as myeloid cell effectors of the osteoblast-driven protumoral response.


Assuntos
Adenocarcinoma/patologia , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Osso e Ossos/patologia , Lectinas/metabolismo , Neoplasias Pulmonares/patologia , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Neutrófilos/patologia , Osteoblastos/patologia , Adenocarcinoma de Pulmão , Animais , Densidade Óssea , Células da Medula Óssea/patologia , Osso e Ossos/metabolismo , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/patologia , Neoplasias Experimentais/patologia , Osteocalcina/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo
6.
Bio Protoc ; 7(11)2017 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-29104894

RESUMO

The host microenvironment plays a prominent role in tumor growth, angiogenesis, invasion, metastasis, and response to therapy. Orthotopic tumor model mimics the natural environment of tumor development and provides an effective approach to investigate tumor pathophysiology and develop therapeutic strategies. This protocol describes the technique involving injection of colorectal cancer cell suspension into the intestinal wall of mice to establish an orthotopic colorectal tumor model.

7.
Cancer Cell ; 31(5): 614-615, 2017 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-28486102

RESUMO

Successful antitumor immunity is thought to require T cell entry into tumors, though mechanisms regulating this process remain unclear. In this issue of Cancer Cell, Spranger et al. indicate that chemokines produced by intratumoral Batf3 dendritic cells are critical for effector T cell recruitment. The findings have implications for immunotherapy.


Assuntos
Linfócitos T/imunologia , Microambiente Tumoral/imunologia , Quimiocinas/imunologia , Células Dendríticas/imunologia , Humanos , Imunoterapia
8.
Am J Cancer Res ; 7(12): 2587-2599, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29312811

RESUMO

Arginine methylation of the epidermal growth factor receptor (meEGFR) increases the binding affinity of EGFR ligands and is reported to have a role in predicting response to anti-EGFR agents. This study investigated the predictive impact of meEGFR in metastatic colorectal cancer (mCRC) patients treated with anti-EGFR agents. Two patient cohorts were evaluated. Cohort 1 consisted of mCRC patients with documented disease progression following anti-EGFR treatment. Circulating tumor cells (CTCs) were isolated and distinguished based on CD45- and Epcam+. Cohort 2 consisted of formalin fixed paraffin-embedded (FFPE) blocks from a prospective cohort. meEGFR in both cohorts was identified by positive staining for me-R198/200 EGFR signal. CTCs were identified in 30 out of 47 cases in cohort 1. Of those 30, meEGFR-CTCs were identified in 19 cases. Mean total meEGFR-CTCs counts was 2.3 (range 0-30) cells per 7.5 ml. There was no association between meEGFR-CTCs and clinic-pathological-molecular features. In RASwt/BRAFwt patients with high levels of meEGFR-CTCs ratio (≥ 0.23) had significantly inferior PFS with anti-EGFR treatment (HR = 3.4, 95% CI 1.5-7.9, P = 0.004). By contrast, high levels of meEGFR in the untreated tumor tissues had no correlation with anti-EGFR treatment duration in cohort 2. Therefore, meEGFR-CTCs may have the potential to serve as a "liquid biopsy" biomarker to predict anti-EGFR treatment efficacy.

9.
Cancer Res ; 76(23): 7049-7058, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27758883

RESUMO

The oncogenic transcription factor Gli1 is a critical effector in the Hedgehog (Hh) pathway, which is necessary for the development and progression of pancreatic ductal adenocarcinoma (PDAC). Although TGFß and K-Ras are known regulators of Gli1 gene transcription in this setting, it is not understood how Gli1 functional activity is regulated. Here, we report the identification of Gli1 as a substrate for the protein arginine N-methyltransferase PRMT1 in PDAC. We found that PRMT1 methylates Gli1 at R597, promoting its transcriptional activity by enhancing the binding of Gli1 to its target gene promoters. Interruption of Gli1 methylation attenuates oncogenic functions of Gli1 and sensitizes PDAC cells to gemcitabine treatment. In human PDAC specimens, the levels of both total Gli1 and methylated Gli1 were correlated positively with PRMT1 protein levels. Notably, PRMT1 regulated Gli1 independently of the canonical Hh pathway as well as the TGFß/Kras-mediated noncanonical Hh pathway, thereby signifying a novel regulatory mechanism for Gli1 transcriptional activity. Taken together, our results identified a new posttranslational modification of Gli1 that underlies its pivotal oncogenic functions in PDAC. Cancer Res; 76(23); 7049-58. ©2016 AACR.


Assuntos
Adenocarcinoma/genética , Carcinogênese/genética , Neoplasias Pancreáticas/genética , Proteína GLI1 em Dedos de Zinco/genética , Adenocarcinoma/patologia , Humanos , Metilação , Neoplasias Pancreáticas/patologia , Transdução de Sinais , Transfecção , Proteína GLI1 em Dedos de Zinco/metabolismo , Neoplasias Pancreáticas
10.
Oncotarget ; 7(35): 57131-57144, 2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27494834

RESUMO

During the process of tumorigenesis, inactivation of tumor suppressors is a critical step. EZH2, a histone methyltransferase, promotes cell growth and migration through catalyzing trimethylation of histone H3 at Lys 27 (H3K27me3) and plays an important role in tumorigenesis. Its expression can be controlled by phosphorylation. However, the regulation of EZH2 activity by tumor suppressor kinase is not well understood. In this study, we show that glycogen synthase kinase 3 beta (GSK3ß) negatively regulates H3K27 trimethylation. We also validate that GSKß physically interacts with EZH2, and their interaction occurs in the cytosol. GSK3ß phosphorylates EZH2 at Ser363 and Thr367 in vitro, and activating GSK3ß upregulates Thr367 phosphorylationin vivo. Cells expressing GSK3ß-non-phosphorylatable mutant EZH2 have higher H3K27 trimethylation and enhanced ability of cell migration and anchorage-independent growth. Inactivation of GSK3ß as measured by its phosphorylation at Ser9 is positively correlated with higher level of H3K27 trimethylation in tumor tissues from breast cancer patients. Our study indicated that GSK3ß phosphorylates EZH2 at Ser363 and Thr367, resulting in reduced H3K27 trimethylation and biological activity of EZH2 in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Metilação de DNA , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Regulação Neoplásica da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/metabolismo , Histonas/química , Catálise , Linhagem Celular Tumoral , Movimento Celular , Citosol/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Mutação , Oncogenes , Fosforilação
11.
Am J Cancer Res ; 6(3): 628-38, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27152240

RESUMO

Pyruvate kinase is a key enzyme in the glycolytic pathway that converts phosphoenolpyruvate to pyruvate, and the M2 isoform of pyruvate kinase (PKM2) is associated with cancer. PKM2 has been reported to function independently of its pyruvate kinase activity, which is crucial for cancer cell proliferation. Moreover, there is growing evidence indicating that dimeric PKM2 is released from tumor cells into the circulation of cancer patients. However, the role of secreted PKM2 in cancer is not well understood. Here, we found that the phosphorylation level of epidermal growth factor receptor (EGFR) significantly increased upon the exposure of cells to the recombinant PKM2 protein. In addition, secreted PKM2 induces EGFR phosphorylation and activates the EGFR downstream signaling in triple-negative breast cancer cells. In contrast, knocking down PKM2 decreased EGFR phosphorylation. Moreover, expression of R399E mutant PKM2, which has been reported to preferentially form a dimer, enhanced EGFR phosphorylation, cellular transformation, and cell proliferation more strongly than the wild-type PKM2. Thus, our study revealed a novel function of extracellular PKM2 in the promoting cancer cell proliferation through EGFR activation.

12.
J Clin Invest ; 125(12): 4529-43, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26571401

RESUMO

Posttranslational modifications to the intracellular domain of the EGFR are known to regulate EGFR functions; however, modifications to the extracellular domain and their effects remain relatively unexplored. Here, we determined that methylation at R198 and R200 of the EGFR extracellular domain by protein arginine methyltransferase 1 (PRMT1) enhances binding to EGF and subsequent receptor dimerization and signaling activation. In a mouse orthotopic colorectal cancer xenograft model, expression of a methylation-defective EGFR reduced tumor growth. Moreover, increased EGFR methylation sustained signaling activation and cell proliferation in the presence of the therapeutic EGFR monoclonal antibody cetuximab. In colorectal cancer patients, EGFR methylation level also correlated with a higher recurrence rate after cetuximab treatment and reduced overall survival. Together, these data indicate that R198/R200 methylation of the EGFR plays an important role in regulating EGFR functionality and resistance to cetuximab treatment.


Assuntos
Cetuximab/farmacologia , Neoplasias do Colo/tratamento farmacológico , Receptores ErbB/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Receptores ErbB/genética , Feminino , Xenoenxertos , Humanos , Metilação/efeitos dos fármacos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteína-Arginina N-Metiltransferases/genética , Proteínas Repressoras/genética , Transdução de Sinais/genética
13.
Cell Res ; 25(2): 225-36, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25601159

RESUMO

Ataxia telangiectasia mutated (ATM) mediates DNA damage response by controling irradiation-induced foci formation, cell cycle checkpoint, and apoptosis. However, how upstream signaling regulates ATM is not completely understood. Here, we show that upon irradiation stimulation, ATM associates with and is phosphorylated by epidermal growth factor receptor (EGFR) at Tyr370 (Y370) at the site of DNA double-strand breaks. Depletion of endogenous EGFR impairs ATM-mediated foci formation, homologous recombination, and DNA repair. Moreover, pretreatment with an EGFR kinase inhibitor, gefitinib, blocks EGFR and ATM association, hinders CHK2 activation and subsequent foci formation, and increases radiosensitivity. Thus, we reveal a critical mechanism by which EGFR directly regulates ATM activation in DNA damage response, and our results suggest that the status of ATM Y370 phosphorylation has the potential to serve as a biomarker to stratify patients for either radiotherapy alone or in combination with EGFR inhibition.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Reparo do DNA , Tirosina/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gefitinibe , Células HeLa , Humanos , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Quinazolinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Radiação Ionizante , Transdução de Sinais/efeitos dos fármacos
14.
Dev Cell ; 30(2): 224-37, 2014 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-25073158

RESUMO

Posttranslational modifications of histones play fundamental roles in many biological functions. Specifically, histone H4-K20 methylation is critical for DNA synthesis and repair. However, little is known about how these functions are regulated by the upstream stimuli. Here, we identify a tyrosine phosphorylation site at Y72 of histone H4, which facilitates recruitment of histone methyltransferases (HMTases), SET8 and SUV4-20H, to enhance its K20 methylation, thereby promoting DNA synthesis and repair. Phosphorylation-defective histone H4 mutant is deficient in K20 methylation, leading to reduced DNA synthesis, delayed cell cycle progression, and decreased DNA repair ability. Disrupting the interaction between epidermal growth factor receptor (EGFR) and histone H4 by Y72 peptide significantly reduced tumor growth. Furthermore, EGFR expression clinically correlates with histone H4-Y72 phosphorylation, H4-K20 monomethylation, and the Ki-67 proliferation marker. These findings uncover a mechanism by which EGFR transduces signal to chromatin to regulate DNA synthesis and repair.


Assuntos
Replicação do DNA , Receptores ErbB/metabolismo , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , Reparo de DNA por Recombinação , Animais , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Fosforilação , Tirosina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Nat Cell Biol ; 13(2): 174-81, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21258366

RESUMO

Epidermal growth factor receptor (EGFR) can undergo post-translational modifications, including phosphorylation, glycosylation and ubiquitylation, leading to diverse physiological consequences and modulation of its biological activity. There is increasing evidence that methylation may parallel other post-translational modifications in the regulation of various biological processes. It is still not known, however, whether EGFR is regulated by this post-translational event. Here, we show that EGFR Arg 1175 is methylated by an arginine methyltransferase, PRMT5. Arg 1175 methylation positively modulates EGF-induced EGFR trans-autophosphorylation at Tyr 1173, which governs ERK activation. Abolishment of Arg 1175 methylation enhances EGF-stimulated ERK activation by reducing SHP1 recruitment to EGFR, resulting in augmented cell proliferation, migration and invasion of EGFR-expressing cells. Therefore, we propose a model in which the regulatory crosstalk between PRMT5-mediated Arg 1175 methylation and EGF-induced Tyr 1173 phosphorylation attenuates EGFR-mediated ERK activation.


Assuntos
Arginina/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Tirosina/metabolismo , Ativação Enzimática , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Células HEK293 , Humanos , Metilação , Fosforilação , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases
16.
ACS Nano ; 4(9): 5414-20, 2010 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-20707382

RESUMO

In this paper, we report on the formation and rupture of Ag nanofilament on planar Ag/TiO2/Pt cells using visual observation. During the forming process, the filament tends to stay very thin. Specifically, it is so thin that it breaks up into a chain of nanospheres (according to Rayleigh instability) right after the formation has been completed. Similar mechanical breakup may also impact vertically stacked cells, causing reliability concerns.

17.
Nucleic Acids Res ; 38(6): 1932-49, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20034954

RESUMO

Epstein-Barr Virus (EBV) DNase (BGLF5) is an alkaline nuclease and has been suggested to be important in the viral life cycle. However, its effect on host cells remains unknown. Serological and histopathological studies implied that EBV DNase seems to be correlated with carcinogenesis. Therefore, we investigate the effect of EBV DNase on epithelial cells. Here, we report that expression of EBV DNase induces increased formation of micronucleus, an indicator of genomic instability, in human epithelial cells. We also demonstrate, using gammaH2AX formation and comet assay, that EBV DNase induces DNA damage. Furthermore, using host cell reactivation assay, we find that EBV DNase expression repressed damaged DNA repair in various epithelial cells. Western blot and quantitative PCR analyses reveal that expression of repair-related genes is reduced significantly in cells expressing EBV DNase. Host shut-off mutants eliminate shut-off expression of repair genes and repress damaged DNA repair, suggesting that shut-off function of BGLF5 contributes to repression of DNA repair. In addition, EBV DNase caused chromosomal aberrations and increased the microsatellite instability (MSI) and frequency of genetic mutation in human epithelial cells. Together, we propose that EBV DNase induces genomic instability in epithelial cells, which may be through induction of DNA damage and also repression of DNA repair, subsequently increases MSI and genetic mutations, and may contribute consequently to the carcinogenesis of human epithelial cells.


Assuntos
Desoxirribonucleases/metabolismo , Instabilidade Genômica , Proteínas Virais/metabolismo , Linhagem Celular , Aberrações Cromossômicas , Quebras de DNA , Dano ao DNA , Reparo do DNA/genética , Células Epiteliais/química , Células Epiteliais/metabolismo , Humanos , Micronúcleos com Defeito Cromossômico , Instabilidade de Microssatélites , Mutação , Biossíntese de Proteínas , Transcrição Gênica , Raios Ultravioleta
18.
Res Microbiol ; 160(1): 71-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19028568

RESUMO

Klebsiella pneumoniae type 3 fimbriae are encoded by mrkABCDF genes which produce the major pilin subunit MrkA, chaperone MrkB, outer membrane usher MrkC, adhesin MrkD and MrkF of unknown function, respectively. RT-PCR analysis demonstrated that the mrkF gene is contained within the mrk operon. Deletion of mrkF in K. pneumoniae CG43 was found to reduce biofilm formation. A higher level of biofilm formation activity was also observed in recombinant Escherichia coli JM109[pmrkABCDF] compared to that observed for JM109[pmrkABCD]. Immunoelectron microscopy analysis of recombinant type 3 fimbriae using anti-MrkA and anti-MrkF antibody-labeled gold particles revealed that MrkF intermittently inserted into the MrkA filament. An interaction between recombinant MrkA and MrkF was demonstrated by co-immunoprecipitation analysis, further supporting the notion that MrkF is a structural component of the fimbriae. Intriguingly, the incorporation of MrkF appeared to decrease fimbrial length but increased activity of autoaggregation and biofilm formation in the bacteria JM109[pmrkABCDF]. This suggested that MrkF may play a role in assembly of the filament.


Assuntos
Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/metabolismo , Klebsiella pneumoniae/genética , Animais , Biofilmes , Escherichia coli/genética , Feminino , Proteínas de Fímbrias/genética , Fímbrias Bacterianas/genética , Deleção de Genes , Genes Bacterianos , Teste de Complementação Genética , Klebsiella pneumoniae/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Família Multigênica , Óperon , RNA Bacteriano/genética , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...