Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Open Forum Infect Dis ; 6(1): ofy338, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30631794

RESUMO

Toxoplasma gondii is a common parasite that infects warm-blooded animals, including humans, and is a foodborne pathogen. We report a case of acute toxoplasmosis in a 76-year-old man after ingestion of the undercooked heart of a white-tailed deer (Odocoileus virginianus) in Tennessee. The patient's adult grandson, who also consumed part of the heart, became ill with nearly identical symptoms, though he did not seek medical care. This case highlights important public health concerns about deer-to-human transmission of Toxoplasma.

2.
Trends Mol Med ; 22(11): 919-921, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27692879

RESUMO

To date, no antiviral agents have been approved for treating Zika virus (ZIKV) infection. Two recent drug-repurposing studies published in Cell Host & Microbe and Nature Medicine demonstrated that screening FDA-approved drugs for antiviral activity is a promising strategy for identifying therapeutics with novel activity against ZIKV infection.


Assuntos
Antivirais/uso terapêutico , Reposicionamento de Medicamentos/métodos , Infecção por Zika virus/tratamento farmacológico , Zika virus/efeitos dos fármacos , Antivirais/farmacologia , Humanos , Transdução de Sinais/efeitos dos fármacos , Infecção por Zika virus/metabolismo
3.
PLoS Comput Biol ; 12(9): e1005074, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27632082

RESUMO

Viruses require host cellular factors for successful replication. A comprehensive systems-level investigation of the virus-host interactome is critical for understanding the roles of host factors with the end goal of discovering new druggable antiviral targets. Gene-trap insertional mutagenesis is a high-throughput forward genetics approach to randomly disrupt (trap) host genes and discover host genes that are essential for viral replication, but not for host cell survival. In this study, we used libraries of randomly mutagenized cells to discover cellular genes that are essential for the replication of 10 distinct cytotoxic mammalian viruses, 1 gram-negative bacterium, and 5 toxins. We herein reported 712 candidate cellular genes, characterizing distinct topological network and evolutionary signatures, and occupying central hubs in the human interactome. Cell cycle phase-specific network analysis showed that host cell cycle programs played critical roles during viral replication (e.g. MYC and TAF4 regulating G0/1 phase). Moreover, the viral perturbation of host cellular networks reflected disease etiology in that host genes (e.g. CTCF, RHOA, and CDKN1B) identified were frequently essential and significantly associated with Mendelian and orphan diseases, or somatic mutations in cancer. Computational drug repositioning framework via incorporating drug-gene signatures from the Connectivity Map into the virus-host interactome identified 110 putative druggable antiviral targets and prioritized several existing drugs (e.g. ajmaline) that may be potential for antiviral indication (e.g. anti-Ebola). In summary, this work provides a powerful methodology with a tight integration of gene-trap insertional mutagenesis testing and systems biology to identify new antiviral targets and drugs for the development of broadly acting and targeted clinical antiviral therapeutics.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas/métodos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Mutagênese Insercional/métodos , Mapas de Interação de Proteínas/efeitos dos fármacos , Biologia de Sistemas/métodos , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Cães , Sistemas de Liberação de Medicamentos , Células Hep G2 , Humanos , Células Madin Darby de Rim Canino , Replicação Viral/genética
4.
Proc Natl Acad Sci U S A ; 112(22): 7073-8, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26038560

RESUMO

Clostridium difficile is the leading cause of hospital-acquired diarrhea in the United States. The two main virulence factors of C. difficile are the large toxins, TcdA and TcdB, which enter colonic epithelial cells and cause fluid secretion, inflammation, and cell death. Using a gene-trap insertional mutagenesis screen, we identified poliovirus receptor-like 3 (PVRL3) as a cellular factor necessary for TcdB-mediated cytotoxicity. Disruption of PVRL3 expression by gene-trap mutagenesis, shRNA, or CRISPR/Cas9 mutagenesis resulted in resistance of cells to TcdB. Complementation of the gene-trap or CRISPR mutants with PVRL3 resulted in restoration of TcdB-mediated cell death. Purified PVRL3 ectodomain bound to TcdB by pull-down. Pretreatment of cells with a monoclonal antibody against PVRL3 or prebinding TcdB to PVRL3 ectodomain also inhibited cytotoxicity in cell culture. The receptor is highly expressed on the surface epithelium of the human colon and was observed to colocalize with TcdB in both an explant model and in tissue from a patient with pseudomembranous colitis. These data suggest PVRL3 is a physiologically relevant binding partner that can serve as a target for the prevention of TcdB-induced cytotoxicity in C. difficile infection.


Assuntos
Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Moléculas de Adesão Celular/metabolismo , Clostridioides difficile/química , Enterotoxinas/toxicidade , Células Epiteliais/metabolismo , Análise de Variância , Anticorpos Monoclonais/metabolismo , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Células CACO-2 , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Colo/metabolismo , Enterotoxinas/metabolismo , Teste de Complementação Genética , Células HeLa , Humanos , Mutagênese Insercional , Nectinas
5.
Virology ; 454-455: 60-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24725932

RESUMO

Previously, we showed that ADAM10 is necessary for HIV-1 replication in primary human macrophages and immortalized cell lines. Silencing ADAM10 expression interrupted the HIV-1 life cycle prior to nuclear translocation of viral cDNA. Furthermore, our data indicated that HIV-1 replication depends on the expression of ADAM15 and γ-secretase, which proteolytically processes ADAM10. Silencing ADAM15 or γ-secretase expression inhibits HIV-1 replication between reverse transcription and nuclear entry. Here, we show that ADAM10 expression also supports replication in CD4(+) T lymphocytes. The intracellular domain (ICD) of ADAM10 associates with the HIV-1 pre-integration complex (PIC) in the cytoplasm and immunoprecipitates and co-localizes with HIV-1 integrase, a key component of PIC. Taken together, our data support a model whereby ADAM15/γ-secretase processing of ADAM10 releases the ICD, which then incorporates into HIV-1 PIC to facilitate nuclear trafficking. Thus, these studies suggest ADAM10 as a novel therapeutic target for inhibiting HIV-1 prior to nuclear entry.


Assuntos
Proteínas ADAM/metabolismo , Transporte Ativo do Núcleo Celular , Secretases da Proteína Precursora do Amiloide/metabolismo , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Substâncias Macromoleculares/metabolismo , Proteínas de Membrana/metabolismo , Integração Viral , Proteína ADAM10 , Células Cultivadas , DNA Viral/metabolismo , Humanos , Imunoprecipitação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Virais/metabolismo
6.
Mol Biotechnol ; 56(5): 429-37, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24477674

RESUMO

We have employed gene-trap insertional mutagenesis to identify candidate genes whose disruption confer phenotypic resistance to lytic infection, in independent studies using 12 distinct viruses and several different cell lines. Analysis of >2,000 virus-resistant clones revealed >1,000 candidate host genes, approximately 20 % of which were disrupted in clones surviving separate infections with 2-6 viruses. Interestingly, there were 83 instances in which the insertional mutagenesis vector disrupted transcripts encoding H/ACA-class and C/D-class small nucleolar RNAs (SNORAs and SNORDs, respectively). Of these, 79 SNORAs and SNORDs reside within introns of 29 genes (predominantly protein-coding), while 4 appear to be independent transcription units. siRNA studies targeting candidate SNORA/Ds provided independent confirmation of their roles in infection when tested against cowpox virus, Dengue Fever virus, influenza A virus, human rhinovirus 16, herpes simplex virus 2, or respiratory syncytial virus. Significantly, eight of the nine SNORA/Ds targeted with siRNAs enhanced cellular resistance to multiple viruses suggesting widespread involvement of SNORA/Ds in virus-host interactions and/or virus-induced cell death.


Assuntos
RNA Nucleolar Pequeno/fisiologia , Replicação Viral/fisiologia , Linhagem Celular , Humanos , Mutagênese Insercional , RNA Interferente Pequeno/genética , RNA Nucleolar Pequeno/genética , Replicação Viral/genética
7.
Infect Immun ; 82(1): 423-32, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24191302

RESUMO

Helicobacter pylori colonizes the human stomach and confers an increased risk for the development of peptic ulceration, noncardia gastric adenocarcinoma, and gastric lymphoma. A secreted H. pylori toxin, VacA, can cause multiple alterations in gastric epithelial cells, including cell death. In this study, we sought to identify host cell factors that are required for VacA-induced cell death. To do this, we analyzed gene trap and short hairpin RNA (shRNA) libraries in AZ-521 human gastric epithelial cells and selected for VacA-resistant clones. Among the VacA-resistant clones, we identified multiple gene trap library clones and an shRNA library clone with disrupted expression of connexin 43 (Cx43) (also known as gap junction protein alpha 1 [GJA1]). Further experiments with Cx43-specific shRNAs confirmed that a reduction in Cx43 expression results in resistance to VacA-induced cell death. Immunofluorescence microscopy experiments indicated that VacA did not colocalize with Cx43. We detected production of the Cx43 protein in AZ-521 cells but not in AGS, HeLa, or RK-13 cells, and correspondingly, AZ-521 cells were the most susceptible to VacA-induced cell death. When Cx43 was expressed in HeLa cells, the cells became more susceptible to VacA. These results indicate that Cx43 is a host cell constituent that contributes to VacA-induced cell death and that variation among cell types in susceptibility to VacA-induced cell death is attributable at least in part to cell type-specific differences in Cx43 production.


Assuntos
Proteínas de Bactérias/fisiologia , Morte Celular/fisiologia , Conexina 43/metabolismo , Células Epiteliais/fisiologia , Helicobacter pylori/fisiologia , Sobrevivência Celular , Células Cultivadas , Mucosa Gástrica/citologia , Humanos , RNA Interferente Pequeno/análise
8.
PLoS One ; 7(10): e46866, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056496

RESUMO

Evidence from multiple studies suggests that Clostridium perfringens ε-toxin is a pore-forming toxin, assembling into oligomeric complexes in the plasma membrane of sensitive cells. In a previous study, we used gene-trap mutagenesis to identify mammalian factors contributing to toxin activity, including caveolin-2 (CAV2). In this study, we demonstrate the importance of caveolin-2 and its interaction partner, caveolin-1 (CAV1), in ε-toxin-induced cytotoxicity. Using CAV2-specific shRNA in a toxin-sensitive human kidney cell line, ACHN, we confirmed that cells deficient in CAV2 exhibit increased resistance to ε-toxin. Similarly, using CAV1-specific shRNA, we demonstrate that cells deficient in CAV1 also exhibit increased resistance to the toxin. Immunoprecipitation of CAV1 and CAV2 from ε-toxin-treated ACHN cells demonstrated interaction of both CAV1 and -2 with the toxin. Furthermore, blue-native PAGE indicated that the toxin and caveolins were components of a 670 kDa protein complex. Although ε-toxin binding was only slightly perturbed in caveolin-deficient cells, oligomerization of the toxin was dramatically reduced in both CAV1- and CAV2-deficient cells. These results indicate that CAV1 and -2 potentiate ε-toxin induced cytotoxicity by promoting toxin oligomerization - an event which is requisite for pore formation and, by extension, cell death.


Assuntos
Toxinas Bacterianas/química , Caveolina 1/metabolismo , Caveolina 2/metabolismo , Multimerização Proteica , Animais , Toxinas Bacterianas/toxicidade , Caveolina 1/deficiência , Caveolina 1/genética , Caveolina 2/deficiência , Caveolina 2/genética , Sobrevivência Celular/efeitos dos fármacos , Cães , Técnicas de Silenciamento de Genes , Humanos , Células Madin Darby de Rim Canino , Estrutura Quaternária de Proteína , RNA Interferente Pequeno/genética
9.
AIDS Res Hum Retroviruses ; 28(10): 1329-39, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22404213

RESUMO

Cellular proteins are essential for human immunodeficiency virus type 1 (HIV-1) replication and may serve as viable new targets for treating infection. Using gene trap insertional mutagenesis, a high-throughput approach based on random inactivation of cellular genes, candidate genes were found that limit virus replication when mutated. Disrupted genes (N=87) conferring resistance to lytic infection with several viruses were queried for an affect on HIV-1 replication by utilizing small interfering RNA (siRNA) screens in TZM-bl cells. Several genes regulating diverse pathways were found to be required for HIV-1 replication, including DHX8, DNAJA1, GTF2E1, GTF2E2, HAP1, KALRN, UBA3, UBE2E3, and VMP1. Candidate genes were independently tested in primary human macrophages, toxicity assays, and/or Tat-dependent ß-galactosidase reporter assays. Bioinformatics analyses indicated that several host factors present in this study participate in canonical pathways and functional processes implicated in prior genome-wide studies. However, the genes presented in this study did not share identity with those found previously. Novel antiviral targets identified in this study should open new avenues for mechanistic investigation.


Assuntos
HIV-1/isolamento & purificação , Mutagênese Insercional/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ativação Viral/genética , Replicação Viral/genética , Linhagem Celular , Células Cultivadas , Feminino , Regulação da Expressão Gênica/genética , HIV-1/fisiologia , Humanos , Masculino , Programas de Rastreamento , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , RNA Viral/genética , Ativação Viral/fisiologia , Replicação Viral/fisiologia
10.
Antivir Chem Chemother ; 22(5): 205-15, 2012 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-22374988

RESUMO

BACKGROUND: Host genes serving potential roles in virus replication may be exploited as novel antiviral targets. METHODS: Small interfering RNA (siRNA)-mediated knockdown of host gene expression was used to validate candidate genes in screens against six unrelated viruses, most importantly influenza. A mouse model of influenza A virus infection was used to evaluate the efficacy of a candidate FDA-approved drug identified in the screening effort. RESULTS: Several genes in the PI3K-AKT-mTOR pathway were found to support broad-spectrum viral replication in vitro by RNA interference. This led to the discovery that everolimus, an mTOR inhibitor, showed in vitro antiviral activity against cowpox, dengue type 2, influenza A, rhino- and respiratory syncytial viruses. In a lethal mouse infection model of influenza A (H1N1 and H5N1) virus infection, everolimus treatment (1 mg/kg/day) significantly delayed death but could not prevent mortality. Fourteen days of treatment was more beneficial in delaying the time to death than treatment for seven days. Pathological findings in everolimus-treated mice showed reduced lung haemorrhage and lung weights in response to infection. CONCLUSIONS: These results provide proof of concept that cellular targets can be identified by gene knockout methods, and highlight the importance of the PI3K-AKT-mTOR pathway in supporting viral infections.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Virus da Influenza A Subtipo H5N1/fisiologia , Mutagênese Insercional , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Everolimo , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Oseltamivir/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribavirina/farmacologia , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
11.
Retrovirology ; 8: 32, 2011 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-21569301

RESUMO

BACKGROUND: Gene trap insertional mutagenesis was used as a high-throughput approach to discover cellular genes participating in viral infection by screening libraries of cells selected for survival from lytic infection with a variety of viruses. Cells harboring a disrupted ADAM10 (A Disintegrin and Metalloprotease 10) allele survived reovirus infection, and subsequently ADAM10 was shown by RNA interference to be important for replication of HIV-1. RESULTS: Silencing ADAM10 expression with small interfering RNA (siRNA) 48 hours before infection significantly inhibited HIV-1 replication in primary human monocyte-derived macrophages and in CD4⁺ cell lines. In agreement, ADAM10 over-expression significantly increased HIV-1 replication. ADAM10 down-regulation did not inhibit viral reverse transcription, indicating that viral entry and uncoating are also independent of ADAM10 expression. Integration of HIV-1 cDNA was reduced in ADAM10 down-regulated cells; however, concomitant 2-LTR circle formation was not detected, suggesting that HIV-1 does not enter the nucleus. Further, ADAM10 silencing inhibited downstream reporter gene expression and viral protein translation. Interestingly, we found that while the metalloprotease domain of ADAM10 is not required for HIV-1 replication, ADAM15 and γ-secretase (which proteolytically release the extracellular and intracellular domains of ADAM10 from the plasma membrane, respectively) do support productive infection. CONCLUSIONS: We propose that ADAM10 facilitates replication at the level of nuclear trafficking. Collectively, our data support a model whereby ADAM10 is cleaved by ADAM15 and γ-secretase and that the ADAM10 intracellular domain directly facilitates HIV-1 nuclear trafficking. Thus, ADAM10 represents a novel cellular target class for development of antiretroviral drugs.


Assuntos
Proteínas ADAM/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Proteínas de Membrana/metabolismo , Replicação Viral , Proteína ADAM10 , Transporte Ativo do Núcleo Celular , Células Cultivadas , HIV-1/fisiologia , Humanos , Macrófagos/virologia , Modelos Biológicos , Mutagênese Insercional , Integração Viral
12.
PLoS One ; 6(3): e17787, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21412435

RESUMO

The Clostridium perfringens ε-toxin is an extremely potent toxin associated with lethal toxemias in domesticated ruminants and may be toxic to humans. Intoxication results in fluid accumulation in various tissues, most notably in the brain and kidneys. Previous studies suggest that the toxin is a pore-forming toxin, leading to dysregulated ion homeostasis and ultimately cell death. However, mammalian host factors that likely contribute to ε-toxin-induced cytotoxicity are poorly understood. A library of insertional mutant Madin Darby canine kidney (MDCK) cells, which are highly susceptible to the lethal affects of ε-toxin, was used to select clones of cells resistant to ε-toxin-induced cytotoxicity. The genes mutated in 9 surviving resistant cell clones were identified. We focused additional experiments on one of the identified genes as a means of validating the experimental approach. Gene expression microarray analysis revealed that one of the identified genes, hepatitis A virus cellular receptor 1 (HAVCR1, KIM-1, TIM1), is more abundantly expressed in human kidney cell lines than it is expressed in human cells known to be resistant to ε-toxin. One human kidney cell line, ACHN, was found to be sensitive to the toxin and expresses a larger isoform of the HAVCR1 protein than the HAVCR1 protein expressed by other, toxin-resistant human kidney cell lines. RNA interference studies in MDCK and in ACHN cells confirmed that HAVCR1 contributes to ε-toxin-induced cytotoxicity. Additionally, ε-toxin was shown to bind to HAVCR1 in vitro. The results of this study indicate that HAVCR1 and the other genes identified through the use of gene-trap mutagenesis and RNA interference strategies represent important targets for investigation of the process by which ε-toxin induces cell death and new targets for potential therapeutic intervention.


Assuntos
Toxinas Bacterianas/toxicidade , Genes/genética , Mamíferos/genética , Mutagênese Insercional/métodos , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Cães , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Ligação Proteica/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Alinhamento de Sequência , Transfecção
13.
Crit Rev Eukaryot Gene Expr ; 16(2): 155-70, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16749895

RESUMO

Viruses are obligate intracellular parasites that rely upon the host cell for activities essential to their life cycles. Gene-trap mutagenesis provides a rapid, genome-wide strategy to identify candidate cellular genes required for virus replication. The candidate genes provide a starting point for mechanistic studies of cellular processes that participate in the virus life cycle and may provide targets for novel antiviral therapies.


Assuntos
Viroses/genética , Animais , Ciclo Celular , Marcação de Genes , Vetores Genéticos , Humanos , Camundongos , Mutagênese , Receptores Virais/genética , Infecções por Reoviridae/genética , Transdução de Sinais , Replicação Viral
14.
J Virol ; 79(18): 11742-51, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16140752

RESUMO

Rab proteins and their effectors facilitate vesicular transport by tethering donor vesicles to their respective target membranes. By using gene trap insertional mutagenesis, we identified Rab9, which mediates late-endosome-to-trans-Golgi-network trafficking, among several candidate host genes whose disruption allowed the survival of Marburg virus-infected cells, suggesting that Rab9 is utilized in Marburg replication. Although Rab9 has not been implicated in human immunodeficiency virus (HIV) replication, previous reports suggested that the late endosome is an initiation site for HIV assembly and that TIP47-dependent trafficking out of the late endosome to the trans-Golgi network facilitates the sorting of HIV Env into virions budding at the plasma membrane. We examined the role of Rab9 in the life cycles of HIV and several unrelated viruses, using small interfering RNA (siRNA) to silence Rab9 expression before viral infection. Silencing Rab9 expression dramatically inhibited HIV replication, as did silencing the host genes encoding TIP47, p40, and PIKfyve, which also facilitate late-endosome-to-trans-Golgi vesicular transport. In addition, silencing studies revealed that HIV replication was dependent on the expression of Rab11A, which mediates trans-Golgi-to-plasma-membrane transport, and that increased HIV Gag was sequestered in a CD63+ endocytic compartment in a cell line stably expressing Rab9 siRNA. Replication of the enveloped Ebola, Marburg, and measles viruses was inhibited with Rab9 siRNA, although the non-enveloped reovirus was insensitive to Rab9 silencing. These results suggest that Rab9 is an important cellular target for inhibiting diverse viruses and help to define a late-endosome-to-plasma-membrane vesicular transport pathway important in viral assembly.


Assuntos
Filoviridae/fisiologia , HIV-1/fisiologia , Vírus do Sarampo/fisiologia , Replicação Viral/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cães , Ebolavirus/genética , Ebolavirus/fisiologia , Filoviridae/genética , Produtos do Gene gag/metabolismo , HIV-1/genética , Humanos , Marburgvirus/genética , Marburgvirus/fisiologia , Vírus do Sarampo/genética , Modelos Biológicos , Mutagênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Viral/genética , RNA Viral/metabolismo , Ratos , Células Vero , Replicação Viral/genética , Proteínas rab de Ligação ao GTP/genética
15.
BMC Cell Biol ; 5(1): 41, 2004 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-15522117

RESUMO

BACKGROUND: Viruses are obligate intracellular parasites that rely upon the host cell for different steps in their life cycles. The characterization of cellular genes required for virus infection and/or cell killing will be essential for understanding viral life cycles, and may provide cellular targets for new antiviral therapies. RESULTS: Candidate genes required for lytic reovirus infection were identified by tagged sequence mutagenesis, a process that permits rapid identification of genes disrupted by gene entrapment. One hundred fifty-one reovirus resistant clones were selected from cell libraries containing 2 x 105 independently disrupted genes, of which 111 contained mutations in previously characterized genes and functionally anonymous transcription units. Collectively, the genes associated with reovirus resistance differed from genes targeted by random gene entrapment in that known mutational hot spots were under represented, and a number of mutations appeared to cluster around specific cellular processes, including: IGF-II expression/signalling, vesicular transport/cytoskeletal trafficking and apoptosis. Notably, several of the genes have been directly implicated in the replication of reovirus and other viruses at different steps in the viral lifecycle. CONCLUSIONS: Tagged sequence mutagenesis provides a rapid, genome-wide strategy to identify candidate cellular genes required for virus infection. The candidate genes provide a starting point for mechanistic studies of cellular processes that participate in the virus lifecycle and may provide targets for novel anti-viral therapies.


Assuntos
Infecções por Reoviridae/genética , Adenocarcinoma/patologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Colo/citologia , Bases de Dados Genéticas , Células Epiteliais/química , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Marcação de Genes/métodos , Genes/genética , Genes Precoces/genética , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Humanos , Células L/química , Células L/metabolismo , Células L/virologia , Camundongos , Mutagênese/genética , Ratos , Reoviridae/genética , Replicação Viral/genética
16.
BMC Cell Biol ; 5: 32, 2004 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-15333144

RESUMO

BACKGROUND: Viruses are obligate intracellular parasites and rely upon the host cell for different steps in their life cycles. The characterization of cellular genes required for virus infection and/or cell killing will be essential for understanding viral life cycles, and may provide cellular targets for new antiviral therapies. RESULTS: A gene entrapment approach was used to identify candidate cellular genes that affect reovirus infection or virus induced cell lysis. Four of the 111 genes disrupted in clones selected for resistance to infection by reovirus type 1 involved the insulin growth factor-2 (IGF-II) pathway, including: the mannose-6-phosphate/IGF2 receptor (Igf2r), a protease associated with insulin growth factor binding protein 5 (Prss11), and the CTCF transcriptional regulator (Ctcf). The disruption of Ctcf, which encodes a repressor of Igf2, was associated with enhanced Igf2 gene expression. Plasmids expressing either the IGF-II pro-hormone or IGF-II without the carboxy terminal extension (E)-peptide sequence independently conferred high levels of cellular resistance to reovirus infection. Forced IGF-II expression results in a block in virus disassembly. In addition, Ctcf disruption and forced Igf2 expression both enabled cells to proliferate in soft agar, a phenotype associated with malignant growth in vivo. CONCLUSION: These results indicate that IGF-II, and by inference other components of the IGF-II signalling pathway, can confer resistance to lytic reovirus infection. This report represents the first use of gene entrapment to identify host factors affecting virus infection. Concomitant transformation observed in some virus resistant cells illustrates a potential mechanism of carcinogenesis associated with chronic virus infection.


Assuntos
Fator de Crescimento Insulin-Like II/genética , Mutagênese , Orthoreovirus de Mamíferos/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Fator de Ligação a CCCTC , Linhagem Celular , Proliferação de Células , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Marcação de Genes , Fator de Crescimento Insulin-Like II/biossíntese , Dados de Sequência Molecular , Mutação , Ratos , Proteínas Repressoras/genética , Transdução de Sinais , Vírion/metabolismo
17.
DNA Cell Biol ; 23(7): 430-41, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15294092

RESUMO

We have utilized growth factors in in vitro and in vivo systems to examine the role of cellular proliferation in reovirus replication. In vitro, proliferating RIE-1 cells can be infected with whole reovirus virions, but are relatively resistant to infection once confluent (Go arrest). It has been shown that TGF-alpha, which signals through the EGF-receptor (EGF-R), is capable of dramatically increasing the number of RIE-1 cells entering the S-phase in the presence of additional serum factors. Stimulation of the EGF-R without serum results in minimal increases in cells entering the S-phase with a restriction in reovirus replication. Therefore, other factors in serum are essential for fully permissive infection. In vivo, we used metallothionein (MT) promoter/enhancer-TGF-alpha transgenic mice to study the effect of cytokine activation on reovirus type 1 infection. Virus replication decreased following oral infection in these transgenic mice at 1 month of age, concordant with increased mucin production. Titers of reovirus obtained from the livers of 1 year old transgenic mice were approximately 10-fold higher than titers obtained in control mice. Taken together, these data indicate that while growth factor activation ultimately leads to an increase in virus infectivity, other factors may be necessary for reovirus replication.


Assuntos
Reoviridae/efeitos dos fármacos , Fator de Crescimento Transformador alfa/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Membrana Celular/metabolismo , Células Cultivadas , Meios de Cultura , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Regulação da Expressão Gênica , Insulina/farmacologia , Intestinos/citologia , Intestinos/virologia , Fígado/virologia , Metalotioneína/genética , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Ratos , Reoviridae/patogenicidade , Reoviridae/fisiologia , Infecções por Reoviridae/genética , Fase S/efeitos dos fármacos , Transdução de Sinais , Fator de Crescimento Transformador alfa/genética , Fator de Crescimento Transformador alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...