Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Virol ; 92(13)2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669830

RESUMO

In humans, homologous to the E6-AP carboxyl terminus (HECT) and regulator of chromosome condensation 1 (RCC1)-like domain-containing protein 5 (HERC5) is an interferon-induced protein that inhibits replication of evolutionarily diverse viruses, including human immunodeficiency virus type 1 (HIV-1). To better understand the origin, evolution, and function of HERC5, we performed phylogenetic, structural, and functional analyses of the entire human small-HERC family, which includes HERC3, HERC4, HERC5, and HERC6. We demonstrated that the HERC family emerged >595 million years ago and has undergone gene duplication and gene loss events throughout its evolution. The structural topology of the RCC1-like domain and HECT domains from all HERC paralogs is highly conserved among evolutionarily diverse vertebrates despite low sequence homology. Functional analyses showed that the human small HERCs exhibit different degrees of antiviral activity toward HIV-1 and that HERC5 provides the strongest inhibition. Notably, coelacanth HERC5 inhibited simian immunodeficiency virus (SIV), but not HIV-1, particle production, suggesting that the antiviral activity of HERC5 emerged over 413 million years ago and exhibits species- and virus-specific restriction. In addition, we showed that both HERC5 and HERC6 are evolving under strong positive selection, particularly blade 1 of the RCC1-like domain, which we showed is a key determinant of antiviral activity. These studies provide insight into the origin, evolution, and biological importance of the human restriction factor HERC5 and the other HERC family members.IMPORTANCE Intrinsic immunity plays an important role as the first line of defense against viruses. Studying the origins, evolution, and functions of proteins responsible for effecting this defense will provide key information about virus-host relationships that can be exploited for future drug development. We showed that HERC5 is one such antiviral protein that belongs to an evolutionarily conserved family of HERCs with an ancient marine origin. Not all vertebrates possess all HERC members, suggesting that different HERCs emerged at different times during evolution to provide the host with a survival advantage. Consistent with this, two of the more recently emerged HERC members, HERC5 and HERC6, displayed strong signatures of having been involved in an ancient evolutionary battle with viruses. Our findings provide new insights into the evolutionary origin and function of the HERC family in vertebrate evolution, identifying HERC5 and possibly HERC6 as important effectors of intrinsic immunity in vertebrates.


Assuntos
Antivirais/metabolismo , Organismos Aquáticos , Evolução Molecular , Infecções por HIV/virologia , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Virais/metabolismo , Infecções por HIV/genética , HIV-1/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Filogenia , Conformação Proteica , Seleção Genética , Proteínas Virais/genética
2.
Hum Mutat ; 35(9): 1072-81, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24863734

RESUMO

Tripartite motif protein 22 (TRIM22) is a novel interferon-induced protein that potently inhibits the replication of evolutionarily diverse viruses, including HIV-1. Altered TRIM22 expression is also associated with diseases, such as multiple sclerosis, cancer, and autoimmunity. The factors that influence TRIM22 expression and antiviral activity are largely unknown. In this study, we adopted an evolution-guided functional approach to identify potential genetic determinants of TRIM22 function. Evolutionary analysis of TRIM22 from mammals spanning >100 million years demonstrated that TRIM22 evolution has been shaped by ancient and variable positive selection. We showed that positive selection is operating on multiple TRIM22 residues that cluster in putative functional regions and that some are predicted to be functionally damaging. Interestingly, the second most prevalent TRIM22 SNP in humans (rs1063303) is located at one of these positively selected sites. We showed that the frequency of rs1063303:G>C varies up to 10-fold between ethnicities and that in some ethnicities SNP rs1063303:G>C is being actively maintained in the population. The SNP rs1063303:G>C variant also had an inverse functional impact where it increased TRIM22 expression and decreased the antiviral activity of TRIM22. Taken together, our data characterize the extensive genetic variation in TRIM22 and identify rs1063303:G>C as a highly prevalent SNP that influences its function.


Assuntos
Adaptação Biológica , Resistência à Doença/genética , Evolução Molecular , Polimorfismo de Nucleotídeo Único , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Viroses/genética , Animais , Linhagem Celular , Humanos , Mamíferos , Antígenos de Histocompatibilidade Menor , Modelos Moleculares , Filogenia , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Proteínas Repressoras/química , Seleção Genética , Proteínas com Motivo Tripartido
3.
Retrovirology ; 8: 95, 2011 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-22093708

RESUMO

BACKGROUND: The identification and characterization of several interferon (IFN)-induced cellular HIV-1 restriction factors, defined as host cellular proteins or factors that restrict or inhibit the HIV-1 life cycle, have provided insight into the IFN response towards HIV-1 infection and identified new therapeutic targets for HIV-1 infection. To further characterize the mechanism underlying restriction of the late stages of HIV-1 replication, we assessed the ability of IFNbeta-induced genes to restrict HIV-1 Gag particle production and have identified a potentially novel host factor called HECT domain and RCC1-like domain-containing protein 5 (HERC5) that blocks a unique late stage of the HIV-1 life cycle. RESULTS: HERC5 inhibited the replication of HIV-1 over multiple rounds of infection and was found to target a late stage of HIV-1 particle production. The E3 ligase activity of HERC5 was required for blocking HIV-1 Gag particle production and correlated with the post-translational modification of Gag with ISG15. HERC5 interacted with HIV-1 Gag and did not alter trafficking of HIV-1 Gag to the plasma membrane. Electron microscopy revealed that the assembly of HIV-1 Gag particles was arrested at the plasma membrane, at an early stage of assembly. The mechanism of HERC5-induced restriction of HIV-1 particle production is distinct from the mechanism underlying HIV-1 restriction by the expression of ISG15 alone, which acts at a later step in particle release. Moreover, HERC5 restricted murine leukemia virus (MLV) Gag particle production, showing that HERC5 is effective in restricting Gag particle production of an evolutionarily divergent retrovirus. CONCLUSIONS: HERC5 represents a potential new host factor that blocks an early stage of retroviral Gag particle assembly. With no apparent HIV-1 protein that directly counteracts it, HERC5 may represent a new candidate for HIV/AIDS therapy.


Assuntos
Produtos do Gene gag/metabolismo , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Montagem de Vírus , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Produtos do Gene gag/genética , HIV-1/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Processamento de Proteína Pós-Traducional , Ubiquitinas/genética , Ubiquitinas/metabolismo
4.
Am J Reprod Immunol ; 80(2): e13019, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30014538

RESUMO

Although anti-retroviral treatments have significantly slowed down the spread of the HIV-1 pandemic, approximately 2 million new infections occur every year. The majority of new infections are in sub-Saharan Africa where rates of infection are much higher in women than men. Young women are disproportionately affected and have higher susceptibility to HIV-1. The complex interactions between HIV-1 and the female genital tract (FGT) and the mechanisms regulating susceptibility in women remain incompletely understood. In this review, we focus on the current understanding of the acute events that occur in the FGT following HIV-1 exposure with a particular focus on the effect of endogenous and exogenous sex hormones on HIV-1 susceptibility. We highlight the contribution of the recent transcriptomic and proteomic studies in providing new insights.


Assuntos
Estrogênios/metabolismo , Genitália Feminina/virologia , Infecções por HIV/transmissão , HIV-1/fisiologia , Mucosa/virologia , Progesterona/metabolismo , Feminino , Genitália Feminina/metabolismo , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , HIV-1/imunologia , Humanos , Imunidade Inata/imunologia , Proteômica/métodos , Biologia de Sistemas/métodos
5.
Am J Reprod Immunol ; 79(1)2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29105931

RESUMO

PROBLEM: Medroxyprogesterone acetate (MPA), a progestin-based hormonal contraceptive designed to mimic progesterone, has been linked to increased human immunodeficiency virus (HIV-1) susceptibility. Genital epithelial cells (GECs) form the mucosal lining of the female genital tract (FGT) and provide the first line of protection against HIV-1. The impact of endogenous sex hormones or MPA on the gene expression profile of GECs has not been comprehensively documented. METHOD OF STUDY: Using microarray analysis, we characterized the transcriptional profile of primary endometrial epithelial cells grown in physiological levels of E2, P4, and MPA. RESULTS: Each hormone treatment altered the gene expression profile of GECs in a unique manner. Interestingly, although MPA is a progestogen, the gene expression profile induced by it was distinct from P4. MPA increased gene expression of genes related to inflammation and cholesterol synthesis linked to innate immunity and HIV-1 susceptibility. CONCLUSION: The analysis of gene expression profiles provides insights into the effects of sex hormones and MPA on GECs and allows us to posit possible mechanisms of the MPA-mediated increase in HIV-1 acquisition.


Assuntos
Anticoncepcionais Femininos/administração & dosagem , Células Epiteliais/efeitos dos fármacos , Genitália Feminina/patologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Acetato de Medroxiprogesterona/administração & dosagem , Mucosa/patologia , Adulto , Células Cultivadas , Colesterol/biossíntese , Anticoncepcionais Femininos/efeitos adversos , Suscetibilidade a Doenças , Células Epiteliais/fisiologia , Feminino , Infecções por HIV/genética , Humanos , Imunidade Inata , Inflamação/genética , Acetato de Medroxiprogesterona/efeitos adversos , Pessoa de Meia-Idade , Cultura Primária de Células , Progesterona/metabolismo , Progestinas/metabolismo , Análise Serial de Tecidos , Transcriptoma
6.
Mol Ther Nucleic Acids ; 3: e187, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25158091

RESUMO

The blood-brain barrier controls the passage of molecules from the blood into the central nervous system (CNS) and is a major challenge for treatment of neurological diseases. Metachromatic leukodystrophy is a neurodegenerative lysosomal storage disease caused by loss of arylsulfatase A (ARSA) activity. Gene therapy via intraventricular injection of a lentiviral vector is a potential approach to rapidly and permanently deliver therapeutic levels of ARSA to the CNS. We present the distribution of integration sites of a lentiviral vector encoding human ARSA (LV-ARSA) in murine brain choroid plexus and ependymal cells, administered via a single intracranial injection into the CNS. LV-ARSA did not exhibit a strong preference for integration in or near actively transcribed genes, but exhibited a strong preference for integration in or near satellite DNA. We identified several genomic hotspots for LV-ARSA integration and identified a consensus target site sequence characterized by two G-quadruplex-forming motifs flanking the integration site. In addition, our analysis identified several other non-B DNA motifs as new factors that potentially influence lentivirus integration, including human immunodeficiency virus type-1 in human cells. Together, our data demonstrate a clinically favorable integration site profile in the murine brain and identify non-B DNA as a potential new host factor that influences lentiviral integration in murine and human cells.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA