Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Nano ; 18(15): 10439-10453, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38567994

RESUMO

The cGAS-STING pathway plays a crucial role in innate immune activation against cancer and infections, and STING agonists based on cyclic dinucleotides (CDN) have garnered attention for their potential use in cancer immunotherapy and vaccines. However, the limited drug-like properties of CDN necessitate an efficient delivery system to the immune system. To address these challenges, we developed an immunostimulatory delivery system for STING agonists. Here, we have examined aqueous coordination interactions between CDN and metal ions and report that CDN mixed with Zn2+ and Mn2+ formed distinctive crystal structures. Further pharmaceutical engineering led to the development of a functional coordination nanoparticle, termed the Zinc-Mn-CDN Particle (ZMCP), produced by a simple aqueous one-pot synthesis. Local or systemic administration of ZMCP exerted robust antitumor efficacy in mice. Importantly, recombinant protein antigens from SARS-CoV-2 can be simply loaded during the aqueous one-pot synthesis. The resulting ZMCP antigens elicited strong cellular and humoral immune responses that neutralized SARS-CoV-2, highlighting ZMCP as a self-adjuvant vaccine platform against COVID-19 and other infectious pathogens. Overall, this work establishes a paradigm for developing translational coordination nanomedicine based on drug-metal ion coordination and broadens the applicability of coordination medicine for the delivery of proteins and other biologics.


Assuntos
Nanopartículas , Neoplasias , Vacinas , Animais , Camundongos , Neoplasias/terapia , Adjuvantes Imunológicos , Imunoterapia/métodos , Nanopartículas/química
2.
J Virol ; 97(12): e0127623, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-37975674

RESUMO

ABSTRACT: Disease progression during SARS-CoV-2 infection is tightly linked to the fate of lung epithelial cells, with severe cases of COVID-19 characterized by direct injury of the alveolar epithelium and an impairment in its regeneration from progenitor cells. The molecular pathways that govern respiratory epithelial cell death and proliferation during SARS-CoV-2 infection, however, remain unclear. We now report a high-throughput CRISPR screen for host genetic modifiers of the survival and proliferation of SARS-CoV-2-infected Calu-3 respiratory epithelial cells. The top four genes identified in our screen encode components of the same type I interferon (IFN-I) signaling complex­IFNAR1, IFNAR2, JAK1, and TYK2. The fifth gene, ACE2, was an expected control encoding the SARS-CoV-2 viral receptor. Surprisingly, despite the antiviral properties of IFN-I signaling, its disruption in our screen was associated with an increase in Calu-3 cell fitness. We validated this effect and found that IFN-I signaling did not sensitize SARS-CoV-2-infected cultures to cell death but rather inhibited the proliferation of surviving cells after the early peak of viral replication and cytopathic effect. We also found that IFN-I signaling alone, in the absence of viral infection, was sufficient to induce this delayed antiproliferative response in both Calu-3 cells and iPSC-derived type 2 alveolar epithelial cells. Together, these findings highlight a cell autonomous antiproliferative response by respiratory epithelial cells to persistent IFN-I signaling during SARS-CoV-2 infection. This response may contribute to the deficient alveolar regeneration that has been associated with COVID-19 lung injury and represents a promising area for host-targeted therapeutic development.


Assuntos
COVID-19 , Células Epiteliais , Interferon Tipo I , Pulmão , Humanos , COVID-19/imunologia , COVID-19/patologia , COVID-19/virologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Interferon Tipo I/imunologia , Pulmão/patologia , Pulmão/virologia , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Linhagem Celular , Proliferação de Células
3.
J Control Release ; 357: 84-93, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36948420

RESUMO

Cyclic dinucleotides (CDNs), as one type of Stimulator of Interferon Genes (STING) pathway agonist, have shown promising results for eliciting immune responses against cancer and viral infection. However, the suboptimal drug-like properties of conventional CDNs, including their short in vivo half-life and poor cellular permeability, compromise their therapeutic efficacy. In this study, we have developed a manganese-silica nanoplatform (MnOx@HMSN) that enhances the adjuvant effects of CDN by achieving synergy with Mn2+ for vaccination against cancer and SARS-CoV-2. MnOx@HMSN with large mesopores were efficiently co-loaded with CDN and peptide/protein antigens. MnOx@HMSN(CDA) amplified the activation of the STING pathway and enhanced the production of type-I interferons and other proinflammatory cytokines from dendritic cells. MnOx@HMSN(CDA) carrying cancer neoantigens elicited robust antitumor T-cell immunity with therapeutic efficacy in two different murine tumor models. Furthermore, MnOx@HMSN(CDA) loaded with SARS-CoV-2 antigen achieved strong and durable (up to one year) humoral immune responses with neutralizing capability. These results demonstrate that MnOx@HMSN(CDA) is a versatile nanoplatform for vaccine applications.


Assuntos
COVID-19 , Neuropatia Hereditária Motora e Sensorial , Nanopartículas , Vacinas , Humanos , Animais , Camundongos , Manganês , Dióxido de Silício , COVID-19/prevenção & controle , SARS-CoV-2 , Imunoterapia
4.
Front Immunol ; 12: 729189, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603303

RESUMO

Several SARS-CoV-2 vaccines have received EUAs, but many issues remain unresolved, including duration of conferred immunity and breadth of cross-protection. Adjuvants that enhance and shape adaptive immune responses that confer broad protection against SARS-CoV-2 variants will be pivotal for long-term protection as drift variants continue to emerge. We developed an intranasal, rationally designed adjuvant integrating a nanoemulsion (NE) that activates TLRs and NLRP3 with an RNA agonist of RIG-I (IVT DI). The combination adjuvant with spike protein antigen elicited robust responses to SARS-CoV-2 in mice, with markedly enhanced TH1-biased cellular responses and high virus-neutralizing antibody titers towards both homologous SARS-CoV-2 and a variant harboring the N501Y mutation shared by B1.1.7, B.1.351 and P.1 variants. Furthermore, passive transfer of vaccination-induced antibodies protected naive mice against heterologous viral challenge. NE/IVT DI enables mucosal vaccination, and has the potential to improve the immune profile of a variety of SARS-CoV-2 vaccine candidates to provide effective cross-protection against future drift variants.


Assuntos
Adjuvantes Imunológicos/farmacologia , Anticorpos Antivirais/imunologia , Vacinas contra COVID-19/imunologia , COVID-19/prevenção & controle , SARS-CoV-2/imunologia , Vacinas Sintéticas/imunologia , Imunidade Adaptativa/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Chlorocebus aethiops , Proteção Cruzada/imunologia , Proteína DEAD-box 58 , Células HEK293 , Humanos , Imunidade Humoral/imunologia , Imunização Passiva , Camundongos , Camundongos Endogâmicos C57BL , Receptores Imunológicos/agonistas , Proteínas Recombinantes/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinação , Células Vero
5.
Immunotherapy ; 11(18): 1527-1531, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31789069

RESUMO

Immune checkpoint inhibitor therapy has become a cornerstone in the management of many oncologic diseases. Although it is well tolerated in most patients, a wide spectrum of adverse events has been described as a result of immune system alteration. We present a case of a woman with metastatic bronchogenic adenocarcinoma who was initially thought to have immune-mediated hepatitis, but eventually discovered to have a rarely described immune-mediated cholangiopathy. Her cholangiopathy appeared to stabilize following ursodeoxycholic acid and tocilizumab after several lines of guideline-directed therapy. Awareness of this unique toxicity following immune checkpoint inhibitor, and potential treatment options may help clinicians manage this rare but serious complication.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/efeitos adversos , Doenças dos Ductos Biliares/induzido quimicamente , Doenças dos Ductos Biliares/tratamento farmacológico , Nivolumabe/efeitos adversos , Ácido Ursodesoxicólico/uso terapêutico , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/terapia , Doenças dos Ductos Biliares/patologia , Doenças dos Ductos Biliares/fisiopatologia , Colagogos e Coleréticos/uso terapêutico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/tratamento farmacológico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/fisiopatologia , Feminino , Humanos , Imunoterapia/efeitos adversos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Pessoa de Meia-Idade , Resultado do Tratamento
6.
J Virol ; 93(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31484747

RESUMO

The endoplasmic reticulum (ER)-resident proteins vesicle-associated membrane protein (VAMP)-associated protein A and B (VAPA and VAPB) have been reported to be necessary for efficient hepatitis C virus (HCV) replication, but the specific mechanisms are not well understood. VAPs are known to recruit lipid transfer proteins to the ER, including oxysterol binding protein (OSBP), which has been previously shown to be necessary for cholesterol delivery to the HCV replication organelle in exchange for phosphatidylinositol 4-phosphate [PI(4)P]. Here, we show that VAPA and VAPB are redundant for HCV infection and that dimerization is not required for their function. In addition, we identify the phosphatidylinositol transfer protein Nir2 as an effector of VAPs to support HCV replication. We propose that Nir2 functions to replenish phosphoinositides at the HCV replication organelle to maintain elevated steady-state levels of PI(4)P, which is removed by OSBP. Thus, Nir2, along with VAPs, OSBP, and the phosphatidylinositol 4-kinase, completes a cycle of phosphoinositide flow between the ER and viral replication organelles to drive ongoing viral replication.IMPORTANCE Hepatitis C virus (HCV) is known for its ability to modulate phosphoinositide signaling pathways for its replication. Elevated levels of phosphatidylinositol 4-phosphate [PI(4)P] in HCV replication organelles (ROs) recruits lipid transfer proteins (LTPs), like oxysterol-binding protein (OSBP). OSBP exchanges PI(4)P with cholesterol, thus removing PI(4)P from the HCV RO. Here, we found that the phosphatidylinositol transfer protein Nir2 acts as an LTP and may replenish PI at the HCV RO by interacting with VAMP-associated proteins (VAPs), enabling continuous viral replication during chronic infection. Therefore, the coordination of OSBP, Nir2, and VAPs completes our understanding of the phosphoinositide cycle between the ER and HCV ROs.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas do Olho/metabolismo , Hepacivirus/fisiologia , Hepatite C Crônica/metabolismo , Hepatite C Crônica/virologia , Proteínas de Membrana/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas R-SNARE/metabolismo , 1-Fosfatidilinositol 4-Quinase/metabolismo , Transporte Biológico , Proteínas de Transporte/metabolismo , Colesterol/metabolismo , Retículo Endoplasmático/metabolismo , Células HEK293 , Hepacivirus/metabolismo , Humanos , Organelas/metabolismo , Organelas/fisiologia , Proteínas de Transporte Vesicular/metabolismo , Replicação Viral/fisiologia
7.
Clin Transl Gastroenterol ; 9(9): 180, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30228268

RESUMO

Immune checkpoint inhibitors (ICPIs) are monoclonal antibodies that target downregulators of the anti-cancer immune response: cytotoxic T-lymphocyte antigen-4, programmed cell death protein-1, and its ligand PD-L1. ICPIs are now approved for the treatment of a wide array of malignancies, with rates of durable responses in the metastatic setting far exceeding what would be expected from conventional chemotherapy. ICPIs have also been associated with rare but serious immune-related adverse events due to over-activation of the immune system that can affect any organ, including the gastrointestinal tract and liver. As the use of ICPIs in oncology continues to increase, ICPI-associated colitis and hepatitis will be encountered frequently by gastroenterologists and hepatologists. This review will focus on the diagnosis and management of ICPI-associated colitis and hepatitis. We will also compare these ICPI-related toxicities with sporadic inflammatory bowel disease and autoimmune liver disease.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Antineoplásicos/efeitos adversos , Antígeno B7-H1/antagonistas & inibidores , Antígeno CTLA-4/antagonistas & inibidores , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Colite/induzido quimicamente , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Doenças Autoimunes/induzido quimicamente , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Colite/diagnóstico , Colite/tratamento farmacológico , Microbioma Gastrointestinal , Humanos , Doenças Inflamatórias Intestinais/induzido quimicamente
8.
mBio ; 8(4)2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28720733

RESUMO

Dengue virus (DENV) is the most common arboviral infection globally, infecting an estimated 390 million people each year. We employed a genome-wide clustered regularly interspaced short palindromic repeat (CRISPR) screen to identify host dependency factors required for DENV propagation and identified the oligosaccharyltransferase (OST) complex as an essential host factor for DENV infection. Mammalian cells express two OSTs containing either STT3A or STT3B. We found that the canonical catalytic function of the OSTs as oligosaccharyltransferases is not necessary for DENV infection, as cells expressing catalytically inactive STT3A or STT3B are able to support DENV propagation. However, the OST subunit MAGT1, which associates with STT3B, is also required for DENV propagation. MAGT1 expression requires STT3B, and a catalytically inactive STT3B also rescues MAGT1 expression, supporting the hypothesis that STT3B serves to stabilize MAGT1 in the context of DENV infection. We found that the oxidoreductase CXXC active site motif of MAGT1 was necessary for DENV propagation, as cells expressing an AXXA MAGT1 mutant were unable to support DENV infection. Interestingly, cells expressing single-cysteine CXXA or AXXC mutants of MAGT1 were able to support DENV propagation. Utilizing the engineered peroxidase APEX2, we demonstrate the close proximity between MAGT1 and NS1 or NS4B during DENV infection. These results reveal that the oxidoreductase activity of the STT3B-containing OST is necessary for DENV infection, which may guide the development of antiviral agents targeting DENV.IMPORTANCE The host oligosaccharyltransferase (OST) complexes have been identified as essential host factors for dengue virus (DENV) replication; however, their functions during DENV infection are unclear. A previous study showed that the canonical OST activity was dispensable for DENV replication, suggesting that the OST complexes serve as scaffolds for DENV replication. However, our work demonstrates that one function of the OST complex during DENV infection is to provide oxidoreductase activity via the OST subunit MAGT1. We also show that MAGT1 associates with DENV NS1 and NS4B during viral infection, suggesting that these nonstructural proteins may be targets of MAGT1 oxidoreductase activity. These results provide insight into the cell biology of DENV infection, which may guide the development of antivirals against DENV.


Assuntos
Vírus da Dengue/fisiologia , Hexosiltransferases/metabolismo , Interações Hospedeiro-Patógeno , Proteínas de Membrana/metabolismo , Oxirredutases/metabolismo , Linhagem Celular , Humanos
9.
Methods Mol Biol ; 1360: 75-85, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26501903

RESUMO

Phosphatidylinositol (PI) and its phosphorylated derivatives, collectively called phosphoinositides, are important second messengers involved in a variety of cellular processes, including cell proliferation, apoptosis, metabolism, and migration. These derivatives are generated by a family of kinases called phosphoinositide lipid kinases (PIKs). Due to the central role of these kinases in signaling pathways, assays for measuring their activity are often used for drug development. Lipid kinase substrates are present in unique membrane environments in vivo and are insoluble in aqueous solutions. Therefore the most important consideration in developing successful lipid kinase assays is the physical state of lipid kinase substrates. Here we describe the preparation of lipid substrates for two major classes of lipid kinases, phosphatidylinositol 3-kinases (PI3Ks) and phosphatidylinositol 4-kinases (PI4Ks). Using PI4Ks as an example, we also provide a detailed protocol for small-scale kinase expression and affinity purification from transiently transfected mammalian cells. For measuring lipid kinase activity we apply a universal bioluminescent ADP detection approach. The approach is compatible with diverse lipid substrates and can be used as a single integrated platform for measuring all classes of lipid and protein kinases.


Assuntos
1-Fosfatidilinositol 4-Quinase/análise , Difosfato de Adenosina/análise , Medições Luminescentes/métodos , Fosfatidilinositol 3-Quinases/análise , Fitas Reagentes , 1-Fosfatidilinositol 4-Quinase/biossíntese , 1-Fosfatidilinositol 4-Quinase/genética , 1-Fosfatidilinositol 4-Quinase/isolamento & purificação , Células HEK293 , Humanos , Indicadores e Reagentes , Luciferases de Vaga-Lume/metabolismo , Micelas , Fosfatidilinositol 3-Quinases/biossíntese , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/isolamento & purificação , Fosfatidilinositóis/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Especificidade por Substrato , Transfecção
10.
J Biol Chem ; 290(47): 28131-28140, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26446786

RESUMO

Upon translocation across the endoplasmic reticulum (ER) membrane, secretory proteins are proteolytically processed to remove their signal peptide by signal peptidase (SPase). This process is critical for subsequent folding, intracellular trafficking, and maturation of secretory proteins. Prokaryotic SPase has been shown to be a promising antibiotic target. In contrast, to date, no eukaryotic SPase inhibitors have been reported. Here we report that introducing a proline immediately following the natural signal peptide cleavage site not only blocks preprotein cleavage but also, in trans, impairs the processing and maturation of co-expressed preproteins in the ER. Specifically, we find that a variant preproinsulin, pPI-F25P, is translocated across the ER membrane, where it binds to the catalytic SPase subunit SEC11A, inhibiting SPase activity in a dose-dependent manner. Similar findings were obtained with an analogous variant of preproparathyroid hormone, demonstrating that inhibition of the SPase does not depend strictly on the sequence or structure of the downstream mature protein. We further show that inhibiting SPase in the ER impairs intracellular processing of viral polypeptides and their subsequent maturation. These observations suggest that eukaryotic SPases (including the human ortholog) are, in principle, suitable therapeutic targets for antiviral drug design.


Assuntos
Retículo Endoplasmático/enzimologia , Proteínas de Membrana/metabolismo , Precursores de Proteínas/metabolismo , Serina Endopeptidases/metabolismo , Células HEK293 , Humanos , Proteólise
11.
Gastroenterology ; 146(5): 1373-85.e1-11, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24512803

RESUMO

BACKGROUND & AIMS: Positive-sense RNA viruses remodel intracellular membranes to generate specialized membrane compartments for viral replication. Several RNA viruses, including poliovirus and hepatitis C virus (HCV), require phosphatidylinositol (PI) 4-kinases for their replication. However, it is not known how PI 4-kinases and their product, PI(4)P, facilitate host membrane reorganization and viral replication. In addition, although the HCV replication compartment, known as the membranous web, is believed to be cholesterol enriched, the mechanisms by which this occurs have not been elucidated. We aimed to identify and characterize a PI 4-kinase effector in HCV replication. METHODS: We used a combination of microscopic and biochemical methods to study HCV replication, web morphology, the distribution of intracellular protein and PI(4)P, along with cholesterol trafficking in HCV-infected cells. PI 4-kinase and oxysterol-binding protein (OSBP) were inhibited using RNA interference or small molecules in cells expressing a full-length genotype 1b replicon or infected with the JFH-1 strain of HCV. RESULTS: OSBP was required for HCV replication and membranous web integrity. OSBP was recruited to membranous webs in a PI 4-kinase-dependent manner, and both these factors were found to regulate cholesterol trafficking to the web. We also found OSBP to be required for poliovirus infection but dispensable for dengue virus. CONCLUSIONS: OSBP is a PI 4-kinase effector in HCV infection, and contributes to the integrity and cholesterol enrichment of the membranous web. OSBP might also be a PI 4-kinase effector in poliovirus infection and could be involved in replication of other viruses that require PI 4-kinases.


Assuntos
1-Fosfatidilinositol 4-Quinase/metabolismo , Membrana Celular/enzimologia , Colesterol/metabolismo , Hepacivirus/enzimologia , Receptores de Esteroides/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , 1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , Transporte Biológico , Membrana Celular/efeitos dos fármacos , Genótipo , Células HEK293 , Células HeLa , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Fenótipo , Fosfatos de Fosfatidilinositol/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Viral/biossíntese , Receptores de Esteroides/genética , Transfecção , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos
12.
PLoS Pathog ; 9(8): e1003513, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935497

RESUMO

Hepatitis C virus (HCV) is a single-stranded RNA virus that replicates on endoplasmic reticulum-derived membranes. HCV particle assembly is dependent on the association of core protein with cellular lipid droplets (LDs). However, it remains uncertain whether HCV assembly occurs at the LD membrane itself or at closely associated ER membranes. Furthermore, it is not known how the HCV replication complex and progeny genomes physically associate with the presumed sites of virion assembly at or near LDs. Using an unbiased proteomic strategy, we have found that Rab18 interacts with the HCV nonstructural protein NS5A. Rab18 associates with LDs and is believed to promote physical interaction between LDs and ER membranes. Active (GTP-bound) forms of Rab18 bind more strongly to NS5A than a constitutively GDP-bound mutant. NS5A colocalizes with Rab18-positive LDs in HCV-infected cells, and Rab18 appears to promote the physical association of NS5A and other replicase components with LDs. Modulation of Rab18 affects genome replication and possibly also the production of infectious virions. Our results support a model in which specific interactions between viral and cellular proteins may promote the physical interaction between membranous HCV replication foci and lipid droplets.


Assuntos
Hepacivirus/fisiologia , Membranas Intracelulares/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Hepatite C/genética , Hepatite C/metabolismo , Humanos , Membranas Intracelulares/virologia , Lipídeos de Membrana/genética , Proteômica , Proteínas não Estruturais Virais/genética , Proteínas rab de Ligação ao GTP/genética
13.
J Hepatol ; 58(6): 1068-73, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23348237

RESUMO

BACKGROUND & AIMS: Hepatitis B virus (HBV) core promoter (CP) mutations have been associated with an increased risk of hepatocellular carcinoma (HCC) in clinical studies. We previously reported that a combination of CP mutations seen in HCC patients, expressed in HBx gene, increased SKP2 (S-phase kinase-associated protein 2) expression, thereby promoting cellular proliferation. Here, we investigate the possible mechanisms by which CP mutations upregulate SKP2. METHODS: We used immunoblotting and ATPlite assay to validate the effect of CP mutations in full-length HBV genome on cell cycle regulator levels and cell proliferation. Activation of SKP2 mRNA was assessed by quantitative real-time PCR in primary human hepatocytes (PHH) and HCC cell lines. Effect of CP mutations on SKP2 promoter activity was determined by luciferase assay. Target regulation of E2F1 on SKP2 was analyzed by siRNAs. RESULTS: CP mutations in full-length HBV genome upregulated SKP2 expression, thereby downregulating cell cycle inhibitors and accelerating cellular proliferation. CP mutations enhanced SKP2 promoter activity but had no effect on SKP2 protein stability. Mapping of the SKP2 promoter identified a region necessary for activation by CP mutations that contains an E2F1 response element. Knocking down E2F1 reduced the effects of CP mutations on SKP2 and cellular proliferation. The effect of CP mutations on E2F1 might be mediated through hyperphosphorylation of RB. CONCLUSIONS: HBV CP mutations enhance SKP2 transcription by activating the E2F1 transcription factor and in turn downregulate cell cycle inhibitors, thus providing a potential mechanism for an association between CP mutations and HCC.


Assuntos
Proliferação de Células , Fator de Transcrição E2F1/fisiologia , Vírus da Hepatite B/genética , Mutação , Regiões Promotoras Genéticas , Proteínas Quinases Associadas a Fase S/genética , Sequência de Bases , Carcinoma Hepatocelular/etiologia , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/etiologia , Dados de Sequência Molecular , Proteínas Quinases Associadas a Fase S/fisiologia , Transativadores/genética , Transcrição Gênica , Regulação para Cima , Proteínas Virais Reguladoras e Acessórias
14.
Transl Res ; 159(6): 421-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22633094

RESUMO

More than 130 million people worldwide are chronically infected with the hepatitis C virus (HCV), which can lead to cirrhosis, liver failure, and hepatocellular carcinoma. Although recently approved HCV NS3-4A protease inhibitors significantly improve treatment response rates, current HCV treatment is still frequently limited by side effects and by the low genetic barrier to viral resistance against direct-acting antiviral agents. A complementary strategy is to target the host cellular factors that support the HCV life cycle. Several studies, including RNA interference screens, demonstrated that HCV depends on dozens, if not hundreds, of cellular proteins to complete its life cycle. A better understanding of the interactions between HCV proteins and host factors may help to identify host targets for antiviral therapy. In this review, we highlight some of the host factors that are particularly attractive targets for the treatment of HCV.


Assuntos
Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Ciclofilinas/fisiologia , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepacivirus/patogenicidade , Hepacivirus/fisiologia , Hepatite C Crônica/fisiopatologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Camundongos , Antígenos de Histocompatibilidade Menor , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Pesquisa Translacional Biomédica , Montagem de Vírus , Internalização do Vírus , Replicação Viral
15.
J Hepatol ; 56(2): 326-33, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21888876

RESUMO

BACKGROUND & AIMS: The precise mechanisms by which IFN exerts its antiviral effect against HCV have not yet been elucidated. We sought to identify host genes that mediate the antiviral effect of IFN-α by conducting a whole-genome siRNA library screen. METHODS: High throughput screening was performed using an HCV genotype 1b replicon, pRep-Feo. Those pools with replicate robust Z scores ≥2.0 entered secondary validation in full-length OR6 replicon cells. Huh7.5.1 cells infected with JFH1 were then used to validate the rescue efficacy of selected genes for HCV replication under IFN-α treatment. RESULTS: We identified and confirmed 93 human genes involved in the IFN-α anti-HCV effect using a whole-genome siRNA library. Gene ontology analysis revealed that mRNA processing (23 genes, p=2.756e-22), translation initiation (nine genes, p=2.42e-6), and IFN signaling (five genes, p=1.00e-3) were the most enriched functional groups. Nine genes were components of U4/U6.U5 tri-snRNP. We confirmed that silencing squamous cell carcinoma antigen recognized by T cells (SART1), a specific factor of tri-snRNP, abrogates IFN-α's suppressive effects against HCV in both replicon cells and JFH1 infectious cells. We further found that SART1 was not IFN-α inducible, and its anti-HCV effector in the JFH1 infectious model was through regulation of interferon stimulated genes (ISGs) with or without IFN-α. CONCLUSIONS: We identified 93 genes that mediate the anti-HCV effect of IFN-α through genome-wide siRNA screening; 23 and nine genes were involved in mRNA processing and translation initiation, respectively. These findings reveal an unexpected role for mRNA processing in generation of the antiviral state, and suggest a new avenue for therapeutic development in HCV.


Assuntos
Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/genética , Interações Hospedeiro-Patógeno/genética , Interferon-alfa/farmacologia , Antígenos de Neoplasias/genética , Linhagem Celular , Biologia Computacional , Genoma Humano , Genômica , Hepacivirus/genética , Hepacivirus/fisiologia , Hepatite C Crônica/virologia , Ensaios de Triagem em Larga Escala , Humanos , Farmacogenética , RNA Interferente Pequeno/genética , Receptor de Interferon alfa e beta/genética , Replicon , Ribonucleoproteínas Nucleares Pequenas/genética
16.
PLoS One ; 6(10): e26300, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22022594

RESUMO

BACKGROUND: Hepatitis C virus (HCV), like other positive-sense RNA viruses, replicates on an altered host membrane compartment that has been called the "membranous web." The mechanisms by which the membranous web are formed from cellular membranes are poorly understood. Several recent RNA interference screens have demonstrated a critical role for the host phosphatidylinositol 4-kinase PI4KA in HCV replication. We have sought to define the function of PI4KA in viral replication. METHODOLOGY/PRINCIPAL FINDINGS: Using a nonreplicative model of membranous web formation, we show that PI4KA silencing leads to aberrant web morphology. Furthermore, we find that PI4KA and its product, phosphatidylinositol 4-phosphate, are enriched on membranous webs and that PI4KA is found in association with NS5A in HCV-infected cells. While the related lipid kinase PI4KB also appears to support HCV replication, it does not interact with NS5A. Silencing of PI4KB does not overtly impair membranous web morphology or phosphatidylinositol 4-phosphate enrichment at webs, suggesting that it acts at a different point in viral replication. Finally, we demonstrate that the aberrant webs induced by PI4KA silencing require the activity of the viral NS3-4A serine protease but not integrity of the host secretory pathway. CONCLUSIONS/SIGNIFICANCE: PI4KA is necessary for the local enrichment of PI 4-phosphate at the HCV membranous web and for the generation of morphologically normal webs. We also show that nonreplicative systems of web formation can be used to order molecular events that drive web assembly.


Assuntos
1-Fosfatidilinositol 4-Quinase/metabolismo , Membrana Celular/metabolismo , Membrana Celular/virologia , Hepacivirus/fisiologia , Interações Hospedeiro-Patógeno , Linhagem Celular Tumoral , Análise por Conglomerados , Retículo Endoplasmático/metabolismo , Inativação Gênica , Complexo de Golgi/metabolismo , Hepatite C/enzimologia , Hepatite C/virologia , Humanos , Fosfatos de Fosfatidilinositol/metabolismo , Poliproteínas/metabolismo , Ligação Proteica , Transporte Proteico , Transdução de Sinais , Proteínas não Estruturais Virais/metabolismo
17.
Gastroenterology ; 141(4): 1412-21, 1421.e1-5, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21704589

RESUMO

BACKGROUND & AIMS: Clinical studies have associated hepatitis B virus core promoter (CP) mutations with an increased risk of hepatocellular carcinoma. The CP region overlaps with the HBV X (HBx) gene, which has been implicated in hepatocarcinogenesis. The cyclin kinase inhibitor p21WAF1/CIP1 is an important regulator of cell cycle progression and proliferation. We determined whether HBx mutants that result from mutations in the CP deregulate p21 and these processes. METHODS: We constructed a series of HBx mutants with changes in the CP region that correspond to A1762T/G1764A (TA), T1753A, T1768A, or a combination of these (combo) and expressed them, along with wild-type HBx under control of its endogenous promoter, in primary human hepatocytes (PHHs) and HepG2 cells. We then analyzed the effects of CP mutations on expression and degradation of p21 and the effects on cell cycle progression and proliferation. RESULTS: The combo mutant decreased levels of p21 and increased cyclin E expression in PHHs and HepG2 cells. The combo mutant, but not HBx with single or double CP mutations, accelerated p21 degradation in HepG2 cells. The combo mutant increased expression of S-phase kinase-associated protein 2 (SKP2) in PHHs and Huh7 cells. Silencing of SKP2 abrogated the effects of CP mutations on p21 expression. The kinetics of p21 expression correlated with changes in cell cycle distribution. The combo mutant accelerated cell cycle progression; p21 overexpression restored G1 arrest. CONCLUSIONS: HBx mutants with changes that correspond to a combination of CP mutations up-regulate SKP2, which then down-regulates p21 via ubiquitin-mediated proteasomal degradation. CP mutations might increase the risk of hepatocellular carcinoma via this pathway.


Assuntos
Carcinoma Hepatocelular/virologia , Transformação Celular Viral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Hepatite B/complicações , Neoplasias Hepáticas/virologia , Mutação , Regiões Promotoras Genéticas , Proteínas Quinases Associadas a Fase S/metabolismo , Transativadores/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Ciclo Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Células Hep G2 , Hepatite B/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Mutagênese Sítio-Dirigida , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Interferência de RNA , Proteínas Quinases Associadas a Fase S/genética , Transdução de Sinais , Fatores de Tempo , Transativadores/metabolismo , Transfecção , Ubiquitinação , Proteínas Virais Reguladoras e Acessórias
18.
Anal Biochem ; 417(1): 97-102, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21704602

RESUMO

Phosphatidylinositol 4-kinases (PI 4-kinases) catalyze the conversion of phosphatidylinositol to phosphatidylinositol 4-phosphate (PtdIns4P). The four known mammalian PI 4-kinases, PI4KA, PI4KB, PI4K2A, and PI4K2B have roles in intracellular lipid and protein trafficking. PI4KA and PI4KB also assist in the replication of several positive-sense RNA viruses. The identification of selective inhibitors of these kinases would be facilitated by assays suitable for high-throughput screening. We describe a homogeneous and nonisotopic assay for PI 4-kinase activity based on the bioluminescent detection of the ADP produced by kinase reactions. We have evaluated this assay with known nonselective inhibitors of PI 4-kinases and show that it performs similar to radiometric assay formats previously described in the literature. In addition, this assay generates Z-factor values of >0.7 for PI4KA in a 384-well format, demonstrating its suitability for high-throughput screening applications.


Assuntos
1-Fosfatidilinositol 4-Quinase/metabolismo , Ensaios Enzimáticos/métodos , 1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , 1-Fosfatidilinositol 4-Quinase/isolamento & purificação , Adenosina/metabolismo , Difosfato de Adenosina/análise , Trifosfato de Adenosina/metabolismo , Androstadienos/farmacologia , Animais , Bovinos , Linhagem Celular , Cromatografia de Afinidade , Dimetil Sulfóxido/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , Marcação por Isótopo , Cinética , Inibidores de Proteínas Quinases/farmacologia , Radiometria , Wortmanina
19.
Biochim Biophys Acta ; 1811(7-8): 476-83, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21601653

RESUMO

Phosphatidylinositol 4-kinase type IIIa (PI4KIIIα) is one of four mammalian PI 4-kinases that catalyzes the first committed step in polyphosphoinositide synthesis. PI4KIIIα has been linked to regulation of ER exit sites and to the synthesis of plasma membrane phosphoinositides and recent studies have also revealed its importance in replication of the Hepatitis C virus in liver. Two isoforms of the mammalian PI4KIIIα have been described and annotated in GenBank: a larger, ~230kDa (isoform 2) and a shorter splice variant containing only the ~97kDa C-terminus that includes the catalytic domain (isoform 1). However, Northern analysis of human tissues and cancer cells showed only a single transcript of ~7.5kb with the exception of the proerythroleukemia line K562, which contained significantly higher level of the 7.5kb transcript along with smaller ones of 2.4, 3.5 and 4.2kb size. Bioinformatic analysis also confirmed the high copy number of PI4KIIIα transcript in K562 cells along with several genes located in the same region in Chr22, including two pseudogenes that cover most exons coding for isoform 1, consistent with chromosome amplification. A panel of polyclonal antibodies raised against peptides within the C-terminal half of PI4KIIIα failed to detect the shorter isoform 1 either in COS-7 cells or K562 cells. Moreover, expression of a cDNA encoding isoform 1 yielded a protein of ~97kDa that showed no catalytic activity and failed to rescue hepatitis C virus replication. These data draw attention to PI4KIIIα as one of the genes found in Chr22q11, a region affected by chromosomal instability, but do not substantiate the existence of a functionally relevant short form of PI4KIIIα.


Assuntos
Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Processamento Alternativo , Animais , Sequência de Bases , Células COS , Chlorocebus aethiops , Cromossomos Humanos Par 22/genética , Primers do DNA/genética , Expressão Gênica , Células HEK293 , Hepacivirus/fisiologia , Humanos , Isoenzimas/classificação , Isoenzimas/genética , Isoenzimas/metabolismo , Células K562 , Antígenos de Histocompatibilidade Menor , Fosfotransferases (Aceptor do Grupo Álcool)/classificação , Pseudogenes , RNA/genética , Replicação Viral/fisiologia
20.
J Hepatol ; 50(4): 705-11, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19231005

RESUMO

BACKGROUND/AIMS: The response rates of HCV infection to interferon therapy vary depending on viral and host factors. We hypothesized that key regulators of the IFN signaling pathway are predictive of treatment outcome. METHODS: We measured the expression of signal transducer and activator of transcription 1 (STAT1) and suppressor of cytokine signaling 3 (SOCS3) in pretreatment liver biopsies. Staining quantitation was compared to treatment outcomes. RESULTS: Forty-nine patients with HCV and 25 patients with HCV/HIV infection treated with peginterferon/ribavirin were analyzed. Pretreatment hepatic SOCS3 expression was higher in non-responders than responders. Genotype 1 responders had similar levels of SOCS3 as genotype 2/3 responders. African Americans (AA) had higher hepatic SOCS3 than non-AA. Pretreatment hepatic SOCS3 was the most powerful independent predictor of sustained virologic response (SVR), even more so than genotype by logistic regression analysis. Failure to achieve SVR and AA race were independently associated with high hepatic SOCS3 levels. The hepatic expression of STAT-1 did not differ between responders and non-responders. CONCLUSIONS: Our data indicate that hepatic SOCS3 is a stronger baseline predictor of antiviral response than viral genotype. Poor response to antiviral therapy in AA may be associated with higher hepatic SOCS3 expression.


Assuntos
Antivirais/uso terapêutico , Infecções por HIV/tratamento farmacológico , Hepacivirus/genética , Hepatite C Crônica/tratamento farmacológico , Fígado/fisiopatologia , Fator de Transcrição STAT1/genética , Proteínas Supressoras da Sinalização de Citocina/genética , Adulto , Biópsia , População Negra/genética , Feminino , Genótipo , Infecções por HIV/complicações , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/complicações , Hepatite C Crônica/patologia , Humanos , Masculino , Valor Preditivo dos Testes , Grupos Raciais/genética , Proteína 3 Supressora da Sinalização de Citocinas , Falha de Tratamento , Resultado do Tratamento , Carga Viral , População Branca/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA