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.
Biomed Pharmacother ; 168: 115698, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37865992

RESUMO

Metastasis is directly linked to poor prognosis of cancer patients and warrants search for effective anti-metastatic drugs. MACC1 is a causal key molecule for metastasis. High MACC1 expression is prognostic for metastasis and poor survival. Here, we developed novel small molecule inhibitors targeting MACC1 expression to impede metastasis formation. We performed a human MACC1 promoter-driven luciferase reporter-based high-throughput screen (HTS; 118.500 compound library) to identify MACC1 transcriptional inhibitors. HTS revealed 1,2,3,4-tetrazolo[1,5-b]pyridazine-based compounds as efficient transcriptional inhibitors of MACC1 expression, able to decrease MACC1-induced cancer cell motility in vitro. Structure-activity relationships identified the essential inhibitory core structure. Best candidates were evaluated for metastasis inhibition in xenografted mouse models demonstrating metastasis restriction. ADMET showed high drug-likeness of these new candidates for cancer therapy. The NFκB pathway was identified as one mode of action targeted by these compounds. Taken together, 1,2,3,4-tetrazolo[1,5-b]pyridazine-based compounds are effective MACC1 inhibitors and pose promising candidates for anti-metastatic therapies particularly for patients with MACC1-overexpressing cancers, that are at high risk to develop metastases. Although further preclinical and clinical development is necessary, these compounds represent important building blocks for an individualized anti-metastatic therapy for solid cancers.


Assuntos
Neoplasias , Transativadores , Animais , Humanos , Camundongos , Regulação Neoplásica da Expressão Gênica , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Regiões Promotoras Genéticas , Transativadores/antagonistas & inibidores
2.
J Orthop Surg Res ; 18(1): 726, 2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37752532

RESUMO

BACKGROUND: Studies have illuminated that long non-coding RNA (lncRNA) influences bone cell differentiation and formation. Nevertheless, whether lncRNA Homeobox D gene cluster antisense growth-associated long noncoding RNA (HAGLR) was implicated in postmenopausal osteoporosis (PMOP) was yet uncertain. PURPOSE: The research was to explore HAGLR's role in the osteogenic differentiation (OD) process of bone marrow mesenchymal stem cells (BMSCs). METHODS: BMSCs were isolated from mouse bone marrow tissues and identified by electron microscope and flow cytometry. HAGLR, microRNA (miR)-182-5p, and homeobox protein A10 (Hoxa10) levels in BMSCs were detected. Mouse BMSC OD process was induced, and calcium deposition and alkaline phosphatase content were analyzed, as well as expressions of runt-related transcription factor 2, osteopontin, and osteocalcin, and cell apoptosis. Bilateral ovaries were resected from mice to construct the ovariectomized model and bone mineral density, maximum bending stress, maximum load, and elastic modulus of the femur were tested, and the femur was histopathologically evaluated. Chondrocyte apoptosis in the articular cartilage of mice was analyzed. Analysis of the interaction of HAGLR, miR-182-5p with Hoxa10 was conducted. RESULTS: HAGLR and Hoxa10 were down-regulated and miR-182-5p was elevated in PMOP patients. During the BMSC OD process, HAGLR and Hoxa10 levels were suppressed, while miR-182-5p was elevated. Promotion of HAGLR or suppression of miR-182-5p accelerated OD of BMSCs. Inhibition of miR-182-5p reversed the inhibitory effect of HAGLR on BMSC OD. In in vivo experiments, up-regulating HAGLR alleviated PMOP, while silencing Hoxa10 reversed the effects of upregulating HAGLR. HAGLR performed as a sponge for miR-182-5p, while miR-182-5p targeted Hoxa10. CONCLUSION: In general, HAGLR boosted the OD process of BMSCs and relieved PMOP via the miR-182-5p/Hoxa10 axis. These data preliminarily reveal the key role of HAGLR in PMOP, and the research results have a certain reference for the treatment of PMOP.


Assuntos
Proteínas Homeobox A10 , Células-Tronco Mesenquimais , MicroRNAs , Osteoporose Pós-Menopausa , RNA Longo não Codificante , Animais , Feminino , Humanos , Camundongos , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Genes Homeobox , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Família Multigênica , Osteoblastos/metabolismo , Osteogênese/genética , Osteoporose Pós-Menopausa/genética , Osteoporose Pós-Menopausa/terapia , Osteoporose Pós-Menopausa/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas Homeobox A10/genética
3.
Cell Signal ; 79: 109886, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33340660

RESUMO

Bladder cancer (BC) is one of the most common tumours of the urinary system and is also known as a highly malignant tumour. In addition to conventional diagnosis and treatment methods, recent research has focused on studying the molecular mechanisms related to BC, in the hope that new, less toxic and effective targeted anticancer drugs and new diagnostic markers can be discovered. It is known that the Wingless (Wnt) signalling pathway and its related genes, proteins and other substances are involved in multiple biological processes of various tumours. Clarifying the contribution of the Wnt signalling pathway in bladder tumours will help establish early diagnosis indicators, develop new therapeutic drugs and evaluate the prognosis for BC. This review aims to summarise previous studies related to BC and the Wnt signalling pathway, with a focus on exploring the participating substances and their mechanisms in the regulation of the Wnt signalling pathway to better determine how to promote new chemotherapeutic drugs, potential therapeutic targets and diagnostic biomarkers.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Via de Sinalização Wnt , Animais , Humanos , Proteínas de Neoplasias/genética , Neoplasias da Bexiga Urinária/genética
4.
Per Med ; 18(1): 9-19, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33052074

RESUMO

Aim: To investigate whether long non-coding RNAs (lncRNAs) can be utilized as molecular biomarkers in predicting the occurrence and progression of chromophobe renal cell carcinoma. Methods & results: Genetic and related clinical traits of chromophobe renal cell carcinoma were downloaded from the Cancer Genome Atlas and used to construct modules using weighted gene coexpression network analysis. In total, 44,889 genes were allocated into 21 coexpression modules depending on intergenic correlation. Among them, the green module was the most significant key module identified by module-trait correlation calculations (R2 = 0.43 and p = 4e-04). Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses demonstrated that genes in the green module were enriched in many pathways. Coexpression, protein-protein interaction networks, screening for differentially expressed genes, and survival analysis were used to select hub lncRNAs. Five hub lncRNAs (TTK, CENPE, KIF2C, BUB1, and RAD51AP1) were selected out. Conclusion: Our findings suggest that the five lncRNAs may act as potential biomarkers for chromophobe renal cell carcinoma progression and prognosis.


Assuntos
Carcinoma de Células Renais/genética , Neoplasias Renais/genética , RNA Longo não Codificante/biossíntese , Biomarcadores Tumorais , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Mapas de Interação de Proteínas , Análise de Sobrevida
5.
Pathogens ; 9(8)2020 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-32722544

RESUMO

Transplantation tolerance is achieved when recipients are unresponsive to donor alloantigen yet mobilize against third-party antigens, including virus. After transplantation, cytomegalovirus (CMV) reactivation in latently-infected transplants reduces allograft viability. To determine if pre-tolerized recipients are resistant to viral dissemination in this setting, we transfused chemically-fixed donor splenocytes (1-ethyl-3- (3'-dimethyl-aminopropyl)-carbo-diimide (ECDI)-treated splenocytes (ECDIsp)) to induce donor antigen tolerance without immunosuppression. In parallel, we implanted donor islet cells to validate operational tolerance. These pre-tolerized recipients were implanted with murine CMV (MCMV) latently-infected donor kidneys (a validated model of CMV latency) to monitor graft inflammation and viral dissemination. Our results indicate that tolerance to donor islets was sustained in recipients after implantation of donor kidneys. In addition, kidney allografts implanted after ECDIsp and islet implantation exhibited low levels of fibrosis and tubulitis. In contrast, kidney cellular and innate immune infiltrates trended higher in the CMV group and exhibited increased markers of CD8+ T cell activation. Tolerance induction was unable to prevent increases in MCMV-specific CD8+ T cells or dissemination of viral IE-1 DNA. Our data suggest that latently-infected allografts are inherently more susceptible to inflammation that is associated with viral dissemination in pre-tolerized recipients. Thus, CMV latently-infected allografts require enhanced strategies to protect allograft integrity and viral spread.

6.
Front Plant Sci ; 10: 1407, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31787996

RESUMO

A highly reproducible plant electrical signal-light-induced bioelectrogenesis (LIB) was obtained by means of periodic illumination/darkness stimulation of broad bean (Vicia faba L.) leaves. By stimulating the same position of the same leaf with different concentrations of NaCl, we observed that the amplitude and waveform of the LIB was correlated with the intensity of stimulation. This method allowed us to link dynamic ion fluxes induced by periodic illumination/darkness to salt stress. The self-referencing ion electrode technique was used to explore the ionic mechanisms of the LIB. Fluxes of H+, Ca2+, K+, and Cl- showed periodic changes under periodic illumination/darkness before and after 50 mM NaCl stimulation. Gray relational analysis was used to analyze correlations between each of these ions and LIB. The results showed that different ions are involved in surface potential changes at different stages under periodic illumination/darkness. The gray relational grade reflected the contribution of each ion to the change in surface potential at a certain time period. The ion fluxes data obtained under periodic illumination/darkness stimulation will contribute to the future development of a dynamic model for interpretation of electrophysiological events in plant cells.

7.
J Gen Virol ; 100(12): 1680-1694, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31647403

RESUMO

CD34+ myeloid lineage progenitor cells are an important reservoir of latent human cytomegalovirus (HCMV), and differentiation to macrophages or dendritic cells (DCs) is known to cause reactivation of latent virus. Due to its species-specificity, murine models have been used to study mouse CMV (MCMV) latency and reactivation in vivo. While previous studies have shown that MCMV genomic DNA can be detected in the bone marrow (BM) of latently infected mice, the identity of these cells has not been defined. Therefore, we sought to identify and enrich for cellular sites of MCMV latency in the BM haematopoietic system, and to explore the potential for establishing an in vitro model for reactivation of latent MCMV. We studied the kinetics and cellular characteristics of acute infection and establishment of latency in the BM of mice. We found that while MCMV can infect a broad range of haematopoietic BM cells (BMCs), latent virus is only detectable in haematopoietic stem cells (HSCs), myeloid progenitor cells, monocytes and DC-enriched cell subsets. Using three separate approaches, MCMV reactivation was detected in association with differentiation into DC-enriched BMCs cultured in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin 4 (IL-4) followed by lipopolysaccharide (LPS) treatment. In summary, we have defined the kinetics and cellular profile of MCMV infection followed by the natural establishment of latency in vivo in the mouse BM haematopoietic system, including the haematopoietic phenotypes of cells that are permissive to acute infection, establish and harbour detectable latent virus, and can be stimulated to reactivate following DC enrichment and differentiation, followed by treatment with LPS.


Assuntos
Células da Medula Óssea/virologia , Diferenciação Celular , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Ativação Viral , Latência Viral , Animais , Biomarcadores , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/virologia , Interações Hospedeiro-Patógeno , Interleucina-4/farmacologia , Cinética , Camundongos , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Células Mieloides/virologia , Tropismo Viral , Replicação Viral
8.
Am J Transplant ; 19(9): 2421-2433, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30947382

RESUMO

Reactivation of latent cytomegalovirus remains an important complication after transplant. Although immunosuppression (IS) has been implicated as a primary cause, we have previously shown that the implantation response of a kidney allograft can lead to early transcriptional activation of latent murine cytomegalovirus (MCMV) genes in an immune-competent host and to MCMV reactivation and dissemination to other organs in a genetically immune-deficient recipient. We now describe a model that allows us to separately analyze the impact of the implantation effect vs that of a clinically relevant IS regimen. Treatment with IS of latently infected mice alone does not induce viral reactivation, but transplant of latently infected allogeneic kidneys combined with IS facilitates MCMV reactivation in the graft and dissemination to other organs. The IS regimen effectively dampens allo-immune inflammatory pathways and depletes recipient anti-MCMV but does not affect ischemia-reperfusion injury pathways. MCMV reactivation similar to that seen in allogeneic transplants combined with also occurs after syngeneic transplants. Thus, our data strongly suggest that while ischemia-reperfusion injury of the implanted graft is sufficient and necessary to initiate transcriptional reactivation of latent MCMV ("first hit"), IS is permissive to the first hit and facilitates dissemination to other organs ("second hit").


Assuntos
Infecções por Citomegalovirus/complicações , Transplante de Rim/efeitos adversos , Muromegalovirus/fisiologia , Insuficiência Renal/cirurgia , Ativação Viral , Animais , Modelos Animais de Doenças , Deleção de Genes , Histonas/metabolismo , Terapia de Imunossupressão , Rim/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Complicações Pós-Operatórias/virologia , Proteômica , Insuficiência Renal/complicações , Traumatismo por Reperfusão , Transplante Homólogo
9.
J Gen Virol ; 97(4): 941-954, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26795571

RESUMO

Reactivation of latent human cytomegalovirus is a significant infectious complication of organ transplantation and current therapies target viral replication once reactivation of latent virus has already occurred. The specific molecular pathways that activate viral gene expression in response to transplantation are not well understood. Our studies aim to identify these factors, with the goal of developing novel therapies that prevent transcriptional reactivation in transplant recipients. Murine cytomegalovirus (MCMV) is a valuable model for studying latency and reactivation of CMV in vivo. We previously demonstrated that transplantation of MCMV-latently infected kidneys into allogeneic recipients induces reactivation of immediate early (IE) gene expression and epigenetic reprogramming of the major immediate early promoter (MIEP) within 48 h. We hypothesize that these events are mediated by activation of signalling pathways that lead to binding of transcription factors to the MIEP, including AP-1 and NF-κB. Here we show that transplantation induces rapid activation of several members of the AP-1 and NF-κB transcription factor family and we demonstrate that canonical NF-κB (p65/p50), the junD component of AP-1, and nucleosome remodelling complexes are recruited to the MIEP following transplantation. Proteomic analysis of recipient plasma and transcriptome analysis of kidney RNA identified five extracellular ligands, including TNF, IL-1ß, IL-18, CD40L and IL-6, and three intracellular signalling pathways associated with reactivation of IE gene expression. Identification of the factors that mediate activation of these signalling pathways may eventually lead to new therapies to prevent reactivation of CMV and its sequelae.


Assuntos
Infecções por Herpesviridae/genética , Proteínas Imediatamente Precoces/genética , Transplante de Rim , Muromegalovirus/genética , NF-kappa B/genética , Fator de Transcrição AP-1/genética , Ativação Viral , Animais , Ligante de CD40/genética , Ligante de CD40/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Interações Hospedeiro-Patógeno , Proteínas Imediatamente Precoces/imunologia , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Muromegalovirus/imunologia , NF-kappa B/imunologia , Nucleossomos/genética , Nucleossomos/imunologia , Regiões Promotoras Genéticas , Proteoma/genética , Proteoma/imunologia , Transdução de Sinais , Fator de Transcrição AP-1/imunologia , Transplante Homólogo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Latência Viral
10.
J Mol Neurosci ; 57(1): 38-47, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25957997

RESUMO

Nur77, together with Nurr1 and NOR-1, constitutes the NR4A subgroup of orphan nuclear receptors and plays critical roles in cell proliferation, differentiation, migration, and apoptosis. Among them, Nur77 is universally well known to contribute to neurite outgrowth. However, information regarding its regulation and possible function in the peripheral nervous system is still limited. In this study, we performed a sciatic nerve injury model in adult rats and detected an increased expression of Nur77 in the sciatic nerve, which was similar to the expression of Oct-6. Immunofluorescence indicated that Nur77 was located in both axons and Schwann cells. In vitro, we observed enhanced expression of Nur77 during the process of both basic fibroblast growth factor (bFGF)-induced Schwann cells differentiation and nerve growth factor (NGF)-induced PC12 cell neurite outgrowth. In vitro and in vivo experiments indicated that inhibiting the function of Nur77 by specific short hairpin RNA could depress Schwann cells myelinization and axons regeneration. Collectively, all these results suggested that upregulation of Nur77 might be involved in Schwann cells differentiation and neurite elongation following sciatic nerve crush.


Assuntos
Neurogênese , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Células de Schwann/metabolismo , Nervo Isquiático/fisiologia , Animais , Células Cultivadas , Masculino , Bainha de Mielina/metabolismo , Regeneração Nervosa , Neuritos/metabolismo , Neuritos/fisiologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Células PC12 , Ratos , Ratos Sprague-Dawley , Células de Schwann/citologia , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Regulação para Cima
11.
J Mol Neurosci ; 56(1): 216-27, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25566957

RESUMO

Nuclear factor (NF)45 (also known as interleukin enhancer-binding factor (ILF)2), is a transcription factor that interacts with NF90 to regulate gene expression. It has long been implicated in the regulation of cell proliferation. However, the role of NF45 in the process of peripheral nervous system regeneration after injury remains poorly understood. Herein, we investigated the spatiotemporal expression of NF45 in a rat sciatic nerve crush model. We detected the up-regulated expression of NF45 in Schwann cell after sciatic nerve crush. What's more, the expression of the cell proliferation marker proliferating cell nuclear antigen (PCNA) exhibited a similar tendency with that of NF45. In cell cultures, we observed increased expression of NF45 during the process of TNF-α-induced Schwann cell proliferation, whereas the protein level of p21 was down-regulated. Interference of NF45 led to enhanced expression of p21 and also impaired proliferation of Schwan cells. Taken together, our data implicated that NF45 was up-regulated in the sciatic nerve after crush, which was associated with proliferation of Schwann cell.


Assuntos
Proliferação de Células , Proteína do Fator Nuclear 45/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Células de Schwann/metabolismo , Nervo Isquiático/metabolismo , Regulação para Cima , Animais , Células Cultivadas , Masculino , Compressão Nervosa , Proteína do Fator Nuclear 45/genética , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Ratos , Ratos Sprague-Dawley , Células de Schwann/fisiologia , Nervo Isquiático/lesões
12.
Neurochem Res ; 39(11): 2105-17, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25103231

RESUMO

Histone deacetylase 4 (HDAC4), a member of the class IIa HDACs subfamily, has emerged as a critical regulator of cell growth, differentiation, and migration in various cell types. It was reported that HDAC4 stimulated colon cell proliferation via repression of p21. Also, HDAC4 contributes to platelet-derived growth factor-BB-induced proliferation and migration of vascular smooth muscle cells. Furthermore, HDAC4 may play an important role in the regulation of neuronal differentiation and survival. However, the role of HDAC4 in the process of peripheral nervous system regeneration after injury remains virtually unknown. Herein, we investigated the spatiotemporal expression of HDAC4 in a rat sciatic nerve crush model. We found that sciatic nerve crush induced up-regulated expression of HDAC4 in Schwann cells. Moreover, the expression of the proliferation marker Ki-67 exhibited a similar tendency with that of HDAC4. In cell cultures, we observed increased expression of HDAC4 during the process of TNF-α-induced Schwann cell proliferation, whereas the protein level of p21 was down-regulated. Interference of HDAC4 led to enhanced expression of p21 and impaired proliferation of Schwan cells. Taken together, our findings implicated that HDAC4 was up-regulated in the sciatic nerve after crush, which was associated with proliferation of Schwann cells.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Histona Desacetilases/metabolismo , Células de Schwann/citologia , Células de Schwann/metabolismo , Nervo Isquiático/lesões , Animais , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Compressão Nervosa/métodos , Regeneração Nervosa/fisiologia , Neurogênese/fisiologia , Ratos Sprague-Dawley , Regulação para Cima
13.
Cell Tissue Res ; 358(2): 277-88, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25030242

RESUMO

Small glutamine-rich tetratricopeptide repeat (TPR)-containing protein alpha (SGTA) is a novel TPR-containing protein involved in various biological processes. However, the expression and roles of SGTA in the central nervous system remain unknown. We have produced an acute spinal cord injury (SCI) model in adult rats and found that SGTA protein levels first significantly increase, reach a peak at day 3 and then gradually return to normal level at day 14 after SCI. These changes are striking in neurons, astrocytes and microglia. Additionally, colocalization of SGTA/active caspase-3 has been detected in neurons and colocalization of SGTA/proliferating cell nuclear antigen has been detected in astrocytes and microglial. In vitro, SGTA depletion by short interfering RNA inhibits astrocyte proliferation and decreases cyclinA and cyclinD1 protein levels. SGTA knockdown also reduces neuronal apoptosis. We speculate that SGTA is involved in biochemical and physiological responses after SCI.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Gliose/etiologia , Neurônios/patologia , Traumatismos da Medula Espinal/complicações , Animais , Biomarcadores/metabolismo , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Imunofluorescência , Técnicas de Silenciamento de Genes , Gliose/patologia , Masculino , Chaperonas Moleculares , Neurônios/metabolismo , Fenótipo , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/patologia
14.
Viruses ; 5(5): 1325-45, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23698401

RESUMO

Cytomegalovirus (CMV) gene expression is repressed in latency due to heterochromatinization of viral genomes. In murine CMV (MCMV) latently infected mice, viral genomes are bound to histones with heterochromatic modifications, to enzymes that mediate these modifications, and to adaptor proteins that may recruit co-repressor complexes. Kinetic analyses of repressor binding show that these repressors are recruited at the earliest time of infection, suggesting that latency may be the default state. Kidney transplantation leads to epigenetic reprogramming of latent viral chromatin and reactivation of immediate early gene expression. Inflammatory signaling pathways, which activate transcription factors that regulate the major immediate early promoter (MIEP), likely mediate the switch in viral chromatin.


Assuntos
Citomegalovirus/fisiologia , Epigênese Genética , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Ativação Viral , Latência Viral , Citomegalovirus/genética , Humanos
15.
J Virol ; 84(7): 3631-43, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20106920

RESUMO

Our previous studies showed that establishment of murine cytomegalovirus (MCMV) latency in vivo is associated with repression of immediate-early gene expression, deacetylation of histones bound to the major immediate-early promoter (MIEP), changes in patterns of methylation of histones, and recruitment of cellular repressors of transcription to the MIEP. Here, we have quantitatively analyzed the kinetics of changes in viral RNA expression, DNA copy number, and recruitment of repressors and activators of transcription to viral promoters during the course of infection. Our results show that changes in viral gene expression correlate with changes in recruitment of RNA polymerase and acetylated histones to viral promoters. Binding of the transcriptional repressors histone deacetylase type 2 (HDAC2), HDAC3, YY1, CBF-1/RBP-Jk, Daxx, and CIR to the MIEP and HDACs to other promoters showed a biphasic pattern: some binding was detectable prior to activation of viral gene expression, then decreased with the onset of transcription and increased again as repression of viral gene expression occurred. Potential binding sites for CBF-1/RBP-Jk and YY1 in the MIEP and for YY1 in the M100 promoter (M100P) were identified by in silico analysis. While recruitment of HDACs was not promoter specific, binding of CBF-1/RBP-Jk and YY1 was restricted to promoters with their cognate sites. Our results suggest that sequences within viral promoters may contribute to establishment of latency through recruitment of transcriptional repressors to these genes. The observation that repressors are bound to the MIEP and other promoters immediately upon infection suggests that latency may be established in some cells very early in infection.


Assuntos
Genes Precoces , Infecções por Herpesviridae/metabolismo , Muromegalovirus/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo , Acetilação , Animais , Feminino , Histona Desacetilases/metabolismo , Histonas/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ativação Transcricional , Fator de Transcrição YY1/metabolismo
16.
Transplantation ; 88(5): 640-5, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19741460

RESUMO

BACKGROUND: Reactivation of cytomegalovirus (CMV) is frequently observed in recipients of solid organs and bone marrow transplants and is associated with increased risk of acute and chronic allograft rejection, opportunistic infection, graft failure, and patient mortality. The molecular mechanisms by which reactivation occurs are not well understood. Previous studies have suggested that tumor necrosis factor (TNF)-alpha, which is induced by allogeneic transplantation, may have a role in reactivation of CMV through activation of nuclear factor kappa-light-chain-enhancer of activated B cells and subsequent transcriptional reactivation of immediate early (ie) gene expression. METHODS AND RESULTS: We have tested the role of TNF-alpha in the reactivation of CMV directly by testing whether TNF-alpha is required to initiate transcription of ie gene expression in a murine model of allogeneic transplantation of kidneys latently infected with mouse CMV. CONCLUSIONS: Our studies show that although TNF-alpha seems to be sufficient, it is not required for initiating transcription of ie gene expression in this model, suggesting that both TNF-alpha-dependent and -independent pathways play an important role in the reactivation of latent CMV infection.


Assuntos
Linfócitos B/virologia , Muromegalovirus/metabolismo , Ativação Transcricional , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linfócitos B/imunologia , Infecções por Citomegalovirus/metabolismo , Regulação da Expressão Gênica , Rejeição de Enxerto , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , NF-kappa B/metabolismo , Transdução de Sinais , Transcrição Gênica
17.
J Virol ; 82(21): 10922-31, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18753203

RESUMO

Human cytomegalovirus (CMV) is a ubiquitous herpesvirus with the ability to establish a lifelong latent infection. The mechanism by which this occurs is not well understood. Regulation of, for example, immediate-early (IE) gene expression is thought to be a critical control point in transcriptional control of the switch between latency and reactivation. Here, we present evidence that supports previous studies showing that the majority of genomes are quiescent with respect to gene expression. To study the possible role of epigenetic factors that may be involved in repression of ie gene expression in latency, we have analyzed changes in the patterns of modifications of histones bound to the major IE promoter (MIEP) in the kidneys of acutely and latently infected mice. Our studies show that, like herpes simplex virus, murine CMV genomes become relatively enriched in histones in latent infection. There are dramatic changes in modifications of histones associated with the MIEP when latency is established: H3 and H4 become hypoacetylated and H3 is hypomethylated at lysine 4, while H3 lysine 9 is hypermethylated in latently infected mice. These changes are accompanied by a relative loss of RNA polymerase and gain of heterochromatin protein 1gamma and Yin-Yang 1 bound to the MIEP. Our studies suggest that, in the majority of cells, CMV establishes a true latent infection, defined as the lack of expression of genes associated with productive infection, and that this occurs through changes in histone modifications and recruitment of transcriptional silencing factors to the MIEP.


Assuntos
DNA Viral/metabolismo , Genes Precoces , Histonas/metabolismo , Muromegalovirus/fisiologia , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo , Latência Viral , Acetilação , Animais , Feminino , Rim/virologia , Metilação , Camundongos , Camundongos Endogâmicos BALB C
18.
J Gen Virol ; 88(Pt 4): 1097-1102, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17374752

RESUMO

We have used a spleen explant model to investigate mechanisms of murine cytomegalovirus latency and reactivation. Induction of immediate-early (ie) gene expression occurs in explants after approximately 9 days in culture and virus reactivation follows induction of ie gene expression with kinetics similar to that of productive infection in vitro. This occurs independently of TNF receptor signalling. Treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine and the histone deacetylase inhibitor trichostatin A results in more rapid induction of ie gene expression and reactivation of virus. Despite these results, which suggest a role for DNA methylation in maintenance of viral latency, we find that the major immediate-early promoter/enhancer is not methylated in latently infected mice. Our results support the hypothesis that latency is maintained by epigenetic control of ie gene expression, and that induction of ie gene expression leads to reactivation of virus, but suggest that these are not controlled by DNA methylation.


Assuntos
Azacitidina/análogos & derivados , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Genes Precoces/efeitos dos fármacos , Ácidos Hidroxâmicos/farmacologia , Muromegalovirus/efeitos dos fármacos , Provírus/efeitos dos fármacos , Ativação Viral/efeitos dos fármacos , Animais , Azacitidina/farmacologia , Metilação de DNA , DNA Viral/química , Decitabina , Inibidores Enzimáticos/farmacologia , Proteínas Imediatamente Precoces/biossíntese , Camundongos , Modelos Animais , Muromegalovirus/genética , Muromegalovirus/fisiologia , Regiões Promotoras Genéticas , Provírus/genética , Provírus/fisiologia , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA