Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nature ; 615(7953): 728-733, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36754086

RESUMEN

The APOBEC3 (A3) proteins are host antiviral cellular proteins that hypermutate the viral genome of diverse viral families. In retroviruses, this process requires A3 packaging into viral particles1-4. The lentiviruses encode a protein, Vif, that antagonizes A3 family members by targeting them for degradation. Diversification of A3 allows host escape from Vif whereas adaptations in Vif enable cross-species transmission of primate lentiviruses. How this 'molecular arms race' plays out at the structural level is unknown. Here, we report the cryogenic electron microscopy structure of human APOBEC3G (A3G) bound to HIV-1 Vif, and the hijacked cellular proteins that promote ubiquitin-mediated proteolysis. A small surface explains the molecular arms race, including a cross-species transmission event that led to the birth of HIV-1. Unexpectedly, we find that RNA is a molecular glue for the Vif-A3G interaction, enabling Vif to repress A3G by ubiquitin-dependent and -independent mechanisms. Our results suggest a model in which Vif antagonizes A3G by intercepting it in its most dangerous form for the virus-when bound to RNA and on the pathway to packaging-to prevent viral restriction. By engaging essential surfaces required for restriction, Vif exploits a vulnerability in A3G, suggesting a general mechanism by which RNA binding helps to position key residues necessary for viral antagonism of a host antiviral gene.


Asunto(s)
Desaminasa APOBEC-3G , VIH-1 , Proteolisis , Productos del Gen vif del Virus de la Inmunodeficiencia Humana , Animales , Humanos , Desaminasa APOBEC-3G/antagonistas & inhibidores , Desaminasa APOBEC-3G/química , Desaminasa APOBEC-3G/metabolismo , Desaminasa APOBEC-3G/ultraestructura , VIH-1/metabolismo , VIH-1/patogenicidad , ARN/química , ARN/metabolismo , Ubiquitina/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/ultraestructura , Microscopía por Crioelectrón , Empaquetamiento del Genoma Viral , Primates/virología
2.
J Virol ; 96(4): e0207121, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34908448

RESUMEN

APOBEC3G (A3G) is a host-encoded cytidine deaminase that potently restricts retroviruses such as HIV-1 and depends on its ability to package into virions. As a consequence of this, HIV-1 protein Vif has evolved to antagonize human A3G by targeting it for ubiquitination and subsequent degradation. There is an ancient arms race between Vif and A3G highlighted by amino acids 128 and 130 in A3G that have evolved under positive selection due to Vif-mediated selective pressure in Old World primates. Nonetheless, not all possible amino acid combinations at these sites have been sampled by nature, and the evolutionary potential of species to resist Vif antagonism is not clear. To explore the evolutionary space of positively selected sites in the Vif-binding region of A3G, we designed a combinatorial mutagenesis screen to introduce all 20 amino acids at sites 128 and 130. Our screen uncovered mutants of A3G with several interesting phenotypes, including loss of antiviral activity and resistance of Vif antagonism. However, HIV-1 Vif exhibited remarkable flexibility in antagonizing A3G 128 and 130 mutants, which significantly reduces viable Vif resistance strategies for hominid primates. Importantly, we find that broadened Vif specificity was conferred through loop 5 adaptations that were required for cross-species adaptation from Old World monkey A3G to hominid A3G. Our evidence suggests that Vif adaptation to novel A3G interfaces during cross-species transmission may train Vif toward broadened specificity that can further facilitate cross-species transmissions and raise the barrier to host resistance. IMPORTANCE APOBEC3G (A3G) is an antiviral protein that potently restricts retroviruses like HIV. In turn, the HIV-1 protein Vif has evolved to antagonize A3G through degradation. Two rapidly evolving sites in A3G confer resistance to unadapted Vif and act as a barrier to cross-species transmission of retroviruses. We recently identified a single amino acid mutation in a simian immunodeficiency virus (SIV) Vif that contributed to the cross-species origins of SIV infecting chimpanzee and, ultimately, the HIV-1 pandemic. This mutation broadened specificity of this Vif to both antagonize the A3G of its host while simultaneously overcoming the A3G barrier in the great apes. In this work, we explore the evolutionary space of human A3G at these rapidly evolving sites to understand if the broadened Vif specificity gained during cross-species transmission confers an advantage to HIV-1 Vif in its host-virus arms race with A3G.


Asunto(s)
Desaminasa APOBEC-3G/antagonistas & inhibidores , VIH-1/fisiología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Desaminasa APOBEC-3G/genética , Adaptación Fisiológica/genética , Aminoácidos , Animales , Infecciones por VIH/transmisión , Infecciones por VIH/virología , VIH-1/genética , Interacciones Microbiota-Huesped , Humanos , Mutación , Primates , Virus de la Inmunodeficiencia de los Simios/genética , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética
3.
Mol Cell Proteomics ; 20: 100132, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34389466

RESUMEN

Structural analysis of host-pathogen protein complexes remains challenging, largely due to their structural heterogeneity. Here, we describe a pipeline for the structural characterization of these complexes using integrative structure modeling based on chemical cross-links and residue-protein contacts inferred from mutagenesis studies. We used this approach on the HIV-1 Vif protein bound to restriction factor APOBEC3G (A3G), the Cullin-5 E3 ring ligase (CRL5), and the cellular transcription factor Core Binding Factor Beta (CBFß) to determine the structure of the (A3G-Vif-CRL5-CBFß) complex. Using the MS-cleavable DSSO cross-linker to obtain a set of 132 cross-links within this reconstituted complex along with the atomic structures of the subunits and mutagenesis data, we computed an integrative structure model of the heptameric A3G-Vif-CRL5-CBFß complex. The structure, which was validated using a series of tests, reveals that A3G is bound to Vif mostly through its N-terminal domain. Moreover, the model ensemble quantifies the dynamic heterogeneity of the A3G C-terminal domain and Cul5 positions. Finally, the model was used to rationalize previous structural, mutagenesis and functional data not used for modeling, including information related to the A3G-bound and unbound structures as well as mapping functional mutations to the A3G-Vif interface. The experimental and computational approach described here is generally applicable to other challenging host-pathogen protein complexes.


Asunto(s)
Desaminasa APOBEC-3G/química , Subunidad beta del Factor de Unión al Sitio Principal/química , Proteínas Cullin/química , Ubiquitina-Proteína Ligasas/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química , Espectrometría de Masas , Modelos Moleculares
4.
Proc Natl Acad Sci U S A ; 116(37): 18607-18612, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451661

RESUMEN

Influenza virus can disseminate from the lungs to the heart in severe infections and can induce cardiac pathology, but this has been difficult to study due to a lack of small animal models. In humans, polymorphisms in the gene encoding the antiviral restriction factor IFN-induced transmembrane protein 3 (IFITM3) are associated with susceptibility to severe influenza, but whether IFITM3 deficiencies contribute to cardiac dysfunction during infection is unclear. We show that IFITM3 deficiency in a new knockout (KO) mouse model increases weight loss and mortality following influenza virus infections. We investigated this enhanced pathogenesis with the A/PR/8/34 (H1N1) (PR8) influenza virus strain, which is lethal in KO mice even at low doses, and observed increased replication of virus in the lungs, spleens, and hearts of KO mice compared with wild-type (WT) mice. Infected IFITM3 KO mice developed aberrant cardiac electrical activity, including decreased heart rate and irregular, arrhythmic RR (interbeat) intervals, whereas WT mice exhibited a mild decrease in heart rate without irregular RR intervals. Cardiac electrical dysfunction in PR8-infected KO mice was accompanied by increased activation of fibrotic pathways and fibrotic lesions in the heart. Infection with a sublethal dose of a less virulent influenza virus strain (A/WSN/33 [H1N1]) resulted in a milder cardiac electrical dysfunction in KO mice that subsided as the mice recovered. Our findings reveal an essential role for IFITM3 in limiting influenza virus replication and pathogenesis in heart tissue and establish IFITM3 KO mice as a powerful model for studying mild and severe influenza virus-induced cardiac dysfunction.


Asunto(s)
Cardiopatías/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/genética , Proteínas de la Membrana/genética , Miocardio/patología , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Electrocardiografía , Fibrosis , Predisposición Genética a la Enfermedad , Corazón/diagnóstico por imagen , Corazón/virología , Cardiopatías/diagnóstico , Cardiopatías/patología , Cardiopatías/virología , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/complicaciones , Gripe Humana/inmunología , Gripe Humana/virología , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Índice de Severidad de la Enfermedad , Replicación Viral/genética , Replicación Viral/inmunología
5.
EMBO Rep ; 18(10): 1740-1751, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28835547

RESUMEN

Interferon-induced transmembrane protein 3 (IFITM3) is a cellular factor that blocks virus fusion with cell membranes. IFITM3 has been suggested to alter membrane curvature and fluidity, though its exact mechanism of action is unclear. Using a bioinformatic approach, we predict IFITM3 secondary structures and identify a highly conserved, short amphipathic helix within a hydrophobic region of IFITM3 previously thought to be a transmembrane domain. Consistent with the known ability of amphipathic helices to alter membrane properties, we show that this helix and its amphipathicity are required for the IFITM3-dependent inhibition of influenza virus, Zika virus, vesicular stomatitis virus, Ebola virus, and human immunodeficiency virus infections. The homologous amphipathic helix within IFITM1 is also required for the inhibition of infection, indicating that IFITM proteins possess a conserved mechanism of antiviral action. We further demonstrate that the amphipathic helix of IFITM3 is required to block influenza virus hemagglutinin-mediated membrane fusion. Overall, our results provide evidence that IFITM proteins utilize an amphipathic helix for inhibiting virus fusion.


Asunto(s)
Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Internalización del Virus , Fenómenos Fisiológicos de los Virus , Biología Computacional , Ebolavirus/fisiología , Células HEK293 , VIH/fisiología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Virus de la Influenza A/fisiología , Estructura Secundaria de Proteína , Virus Zika/fisiología
6.
PLoS Pathog ; 11(8): e1005095, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26263374

RESUMEN

Interferon (IFN)-induced transmembrane protein 3 (IFITM3) is a cell-intrinsic factor that limits influenza virus infections. We previously showed that IFITM3 degradation is increased by its ubiquitination, though the ubiquitin ligase responsible for this modification remained elusive. Here, we demonstrate that the E3 ubiquitin ligase NEDD4 ubiquitinates IFITM3 in cells and in vitro. This IFITM3 ubiquitination is dependent upon the presence of a PPxY motif within IFITM3 and the WW domain-containing region of NEDD4. In NEDD4 knockout mouse embryonic fibroblasts, we observed defective IFITM3 ubiquitination and accumulation of high levels of basal IFITM3 as compared to wild type cells. Heightened IFITM3 levels significantly protected NEDD4 knockout cells from infection by influenza A and B viruses. Similarly, knockdown of NEDD4 in human lung cells resulted in an increase in steady state IFITM3 and a decrease in influenza virus infection, demonstrating a conservation of this NEDD4-dependent IFITM3 regulatory mechanism in mouse and human cells. Consistent with the known association of NEDD4 with lysosomes, we demonstrate for the first time that steady state turnover of IFITM3 occurs through the lysosomal degradation pathway. Overall, this work identifies the enzyme NEDD4 as a new therapeutic target for the prevention of influenza virus infections, and introduces a new paradigm for up-regulating cellular levels of IFITM3 independently of IFN or infection.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Gripe Humana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Línea Celular , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Microscopía Fluorescente , Datos de Secuencia Molecular , Ubiquitina-Proteína Ligasas Nedd4 , ARN Interferente Pequeño , Transfección , Ubiquitinación
7.
J Biol Chem ; 289(17): 11986-11992, 2014 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-24627473

RESUMEN

Interferon-inducible transmembrane protein 3 (IFITM3) is essential for innate defense against influenza virus in mice and humans. IFITM3 localizes to endolysosomes where it prevents virus fusion, although mechanisms controlling its trafficking to this cellular compartment are not fully understood. We determined that both mouse and human IFITM3 are phosphorylated by the protein-tyrosine kinase FYN on tyrosine 20 (Tyr(20)) and that mouse IFITM3 is also phosphorylated on the non-conserved Tyr(27). Phosphorylation led to a cellular redistribution of IFITM3, including plasma membrane accumulation. Mutation of Tyr(20) caused a similar redistribution of IFITM3 and resulted in decreased antiviral activity against influenza virus, whereas Tyr(27) mutation of mouse IFITM3 showed minimal effects on localization or activity. Using FYN knockout cells, we also found that IFITM3 phosphorylation is not a requirement for its antiviral activity. Together, these results indicate that Tyr(20) is part of an endocytosis signal that can be blocked by phosphorylation or by mutation of this residue. Further mutagenesis narrowed this endocytosis-controlling region to four residues conforming to a YXXΦ (where X is any amino acid and Φ is Val, Leu, or Ile) endocytic motif that, when transferred to CD4, resulted in its internalization from the cell surface. Additionally, we found that phosphorylation of IFITM3 by FYN and mutagenesis of Tyr(20) both resulted in decreased IFITM3 ubiquitination. Overall, these results suggest that modification of Tyr(20) may serve in a cellular checkpoint controlling IFITM3 trafficking and degradation and demonstrate the complexity of posttranslational regulation of IFITM3.


Asunto(s)
Endocitosis , Proteínas de la Membrana/fisiología , Ubiquitinación , Secuencia de Aminoácidos , Animales , Células Cultivadas , Células HEK293 , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Fosforilación , Tirosina/metabolismo
8.
Am J Physiol Lung Cell Mol Physiol ; 308(11): L1136-44, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25840995

RESUMEN

As the eighth leading cause of annual mortality in the USA, influenza A viruses are a major public health concern. In 20% of patients, severe influenza progresses to acute lung injury (ALI). However, pathophysiological mechanisms underlying ALI development are poorly defined. We reported that, unlike wild-type (WT) C57BL/6 controls, influenza A virus-infected mice that are heterozygous for the F508del mutation in the cystic fibrosis transmembrane conductance regulator (HETs) did not develop ALI. This effect was associated with higher IL-6 and alveolar macrophages (AMs) at 6 days postinfection (d.p.i.) in HET bronchoalveolar lavage fluid (BALF). In the present study, we found that HET AMs were an important source of IL-6 at 6 d.p.i. Infection also induced TGF-ß production by HET but not WT mice at 2 d.p.i. TGF-ß neutralization at 2 d.p.i. (TGF-N) significantly reduced BALF IL-6 in HETs at 6 d.p.i. Neither TGF-N nor IL-6 neutralization at 4 d.p.i. (IL-6-N) altered postinfection weight loss or viral replication in either mouse strain. However, both treatments increased influenza A virus-induced hypoxemia, pulmonary edema, and lung dysfunction in HETs to WT levels at 6 d.p.i. TGF-N and IL-6-N did not affect BALF AM and neutrophil numbers but attenuated the CXCL-1/keratinocyte chemokine response in both strains and reduced IFN-γ production in WT mice. Finally, bone marrow transfer experiments showed that HET stromal and myeloid cells are both required for protection from ALI in HETs. These findings indicate that TGF-ß-dependent production of IL-6 by AMs later in infection prevents ALI development in influenza A virus-infected HET mice.


Asunto(s)
Lesión Pulmonar Aguda/virología , Virus de la Influenza A/inmunología , Interleucina-6/fisiología , Infecciones por Orthomyxoviridae/inmunología , Factor de Crecimiento Transformador beta/fisiología , Lesión Pulmonar Aguda/inmunología , Animales , Líquido del Lavado Bronquioalveolar , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Inmunidad Innata , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos CFTR , Eliminación de Secuencia
9.
BMC Biol ; 12: 91, 2014 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-25371237

RESUMEN

BACKGROUND: Palmitoylation is a 16-carbon lipid post-translational modification that increases protein hydrophobicity. This form of protein fatty acylation is emerging as a critical regulatory modification for multiple aspects of cellular interactions and signaling. Despite recent advances in the development of chemical tools for the rapid identification and visualization of palmitoylated proteins, the palmitoyl proteome has not been fully defined. Here we sought to identify and compare the palmitoylated proteins in murine fibroblasts and dendritic cells. RESULTS: A total of 563 putative palmitoylation substrates were identified, more than 200 of which have not been previously suggested to be palmitoylated in past proteomic studies. Here we validate the palmitoylation of several new proteins including Toll-like receptors (TLRs) 2, 5 and 10, CD80, CD86, and NEDD4. Palmitoylation of TLR2, which was uniquely identified in dendritic cells, was mapped to a transmembrane domain-proximal cysteine. Inhibition of TLR2 S-palmitoylation pharmacologically or by cysteine mutagenesis led to decreased cell surface expression and a decreased inflammatory response to microbial ligands. CONCLUSIONS: This work identifies many fatty acylated proteins involved in fundamental cellular processes as well as cell type-specific functions, highlighting the value of examining the palmitoyl proteomes of multiple cell types. S-palmitoylation of TLR2 is a previously unknown immunoregulatory mechanism that represents an entirely novel avenue for modulation of TLR2 inflammatory activity.


Asunto(s)
Lipoilación , Proteómica/métodos , Receptor Toll-Like 2/metabolismo , Acilación , Animales , Antígeno B7-2/genética , Antígeno B7-2/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Química Clic , Biología Computacional , Células Dendríticas/metabolismo , Fibroblastos/metabolismo , Células HEK293 , Humanos , Interferón-alfa/metabolismo , Ligandos , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Procesamiento Proteico-Postraduccional , Proteoma/genética , Proteoma/metabolismo , Receptor Toll-Like 2/genética
10.
J Virol ; 87(17): 9923-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804635

RESUMEN

The interferon-induced transmembrane proteins (IFITMs) restrict infection by numerous viruses, yet the importance and regulation of individual isoforms remains unclear. Here, we report that murine IFITM1 (mIFITM1) is palmitoylated on one nonconserved cysteine and three conserved cysteines that are required for anti-influenza A virus activity. Additionally, palmitoylation of mIFITM1 regulates protein stability by preventing proteasomal degradation, and modification of the nonconserved cysteine at the mIFITM1 C terminus supports an intramembrane topology with mechanistic implications.


Asunto(s)
Antígenos de Diferenciación/química , Antígenos de Diferenciación/fisiología , Virus de la Influenza A/patogenicidad , Secuencia de Aminoácidos , Animales , Antígenos de Diferenciación/genética , Secuencia Conservada , Cisteína/química , Cisteína/genética , Células HEK293 , Humanos , Virus de la Influenza A/fisiología , Lipoilación , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Estabilidad Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Internalización del Virus
11.
Viruses ; 12(4)2020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-32235597

RESUMEN

The APOBEC3 family of cytidine deaminases are an important part of the host innate immune defense against endogenous retroelements and retroviruses like Human Immunodeficiency Virus (HIV). APOBEC3H (A3H) is the most polymorphic of the human APOBEC3 genes, with four major haplotypes circulating in the population. Haplotype II is the only antivirally-active variant of A3H, while the majority of the population possess independently destabilizing polymorphisms present in haplotype I (R105G) and haplotypes III and IV (N15del). In this paper, we show that instability introduced by either polymorphism is positively correlated with degradative ubiquitination, while haplotype II is protected from this modification. Inhibiting ubiquitination by mutating all of the A3H lysines increased the expression of haplotypes III and IV, but these stabilized forms of haplotype III and IV had a strict nuclear localization, and did not incorporate into virions, nor exhibit antiviral activity. Fusion chimeras with haplotype II allowed for stabilization, cytoplasmic retention, and packaging of the N15del-containing haplotype III, but the haplotype III component of these chimeras was unable to restrict HIV-1 on its own. Thus, the evolutionary loss of A3H activity in many humans involves functional deficiencies independent of protein stability.


Asunto(s)
Aminohidrolasas/genética , Polimorfismo Genético , Ubiquitinación , Virosis/genética , Virosis/virología , Línea Celular , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Haplotipos , Humanos , Estabilidad Proteica , Ensamble de Virus , Virosis/inmunología , Fenómenos Fisiológicos de los Virus
12.
Cell Host Microbe ; 26(6): 739-747.e4, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31830442

RESUMEN

Primate lentiviruses encode a Vif protein that counteracts the host antiviral APOBEC3 (A3) family members. The adaptation of Vif to species-specific A3 determinants is a critical event that allowed the spillover of a lentivirus from monkey reservoirs to chimpanzees and subsequently to humans, which gave rise to HIV-1 and the acquired immune deficiency syndrome (AIDS) pandemic. How Vif-A3 protein interactions are remodeled during evolution is unclear. Here, we report a 2.94 Å crystal structure of the Vif substrate receptor complex from simian immunodeficiency virus isolated from red-capped mangabey (SIVrcm). The structure of the SIVrcm Vif complex illuminates the stage of lentiviral Vif evolution that is immediately prior to entering hominid primates. Structure-function studies reveal the adaptations that allowed SIVrcm Vif to antagonize hominid A3G. These studies show a partitioning between an evolutionarily dynamic specificity determinant and a conserved protein interacting surface on Vif that enables adaptation while maintaining protein interactions required for potent A3 antagonism.


Asunto(s)
Productos del Gen vif , Virus de la Inmunodeficiencia de los Simios , Desaminasa APOBEC-3G/metabolismo , Síndrome de Inmunodeficiencia Adquirida , Animales , Cercocebus , Cristalografía , Evolución Molecular , Productos del Gen vif/química , Productos del Gen vif/genética , VIH-1/genética , VIH-1/metabolismo , Hominidae , Interacciones Huésped-Patógeno , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Enfermedades de los Monos/virología , Pan troglodytes , Primates , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/ultraestructura
13.
Viruses ; 7(6): 3035-52, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-26075508

RESUMEN

Interferon induced transmembrane proteins (IFITMs) found in vertebrates restrict infections by specific viruses. IFITM3 is known to be essential for restriction of influenza virus infections in both mice and humans. Vertebrate IFITMs are hypothesized to have derived from a horizontal gene transfer from bacteria to a primitive unicellular eukaryote. Since bacterial IFITMs share minimal amino acid identity with human IFITM3, we hypothesized that examination of bacterial IFITMs in human cells would provide insight into the essential characteristics necessary for antiviral activity of IFITMs. We examined IFITMs from Mycobacterium avium and Mycobacterium abscessus for potential antiviral activity. Both of these IFITMs conferred a moderate level of resistance to influenza virus in human cells, identifying them as functional homologues of IFITM3. Analysis of sequence elements shared by bacterial IFITMs and IFITM3 identified two hydrophobic domains, putative S-palmitoylation sites, and conserved phenylalanine residues associated with IFITM3 interactions, which are all necessary for IFITM3 antiviral activity. We observed that, like IFITM3, bacterial IFITMs were S-palmitoylated, albeit to a lesser degree. We also demonstrated the ability of a bacterial IFITM to co-immunoprecipitate with IFITM3 suggesting formation of a complex, and also visualized strong co-localization of bacterial IFITMs with IFITM3. However, the mycobacterial IFITMs lack the endocytic-targeting motif conserved in vertebrate IFITM3. As such, these bacterial proteins, when expressed alone, had diminished colocalization with cathepsin B-positive endolysosomal compartments that are the primary site of IFITM3-dependent influenza virus restriction. Though the precise evolutionary origin of vertebrate IFITMs is not known, our results support a model whereby transfer of a bacterial IFITM gene to eukaryotic cells may have provided a selective advantage against viral infection that was refined through the course of vertebrate evolution to include more robust signals for S-palmitoylation and localization to sites of endocytic virus trafficking.


Asunto(s)
Antivirales/metabolismo , Proteínas Bacterianas/metabolismo , Expresión Génica , Inductores de Interferón/metabolismo , Mycobacterium/inmunología , Orthomyxoviridae/inmunología , Proteínas Bacterianas/genética , Línea Celular , Secuencia Conservada , Endosomas/química , Células Epiteliales/química , Células Epiteliales/inmunología , Células Epiteliales/virología , Humanos , Lipoilación , Mycobacterium/genética , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
14.
J Control Release ; 203: 140-9, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25687308

RESUMEN

Alveolar type II (ATII) respiratory epithelial cells are essential to normal lung function. They may be also central to the pathogenesis of diseases such as acute lung injury, pulmonary fibrosis, and pulmonary adenocarcinoma. Hence, ATII cells are important therapeutic targets. However, effective ATII cell-specific drug delivery in vivo requires carriers of an appropriate size, which can cross the hydrophobic alveolar surfactant film and polar aqueous layer overlying ATII cells, and be taken up without inducing ATII cell dysfunction, pulmonary inflammation, lung damage, or excessive systemic spread and side-effects. We have developed lipoplexes as a versatile nanoparticle carrier system for drug/RNA delivery. To optimize their pulmonary localization and ATII cell specificity, lipoplexes were conjugated to an antibody directed against the ATII cell-specific antigen surfactant protein-C (SP-C) then administered to C57BL/6 mice via the nares. Intranasally-administered, anti-SP-C-conjugated lipoplexes targeted mouse ATII cells with >70% specificity in vivo, were retained within ATII cells for at least 48h, and did not accumulate at significant levels in other lung cell types or viscera. 48h after treatment with anti-SP-C-conjugated lipoplexes containing the test microRNA miR-486, expression of mature miR-486 was approximately 4-fold higher in ATII cells than whole lung by qRT-PCR, and was undetectable in other viscera. Lipoplexes induced no weight loss, hypoxemia, lung dysfunction, pulmonary edema, or pulmonary inflammation over a 6-day period. These findings indicate that ATII cell-targeted lipoplexes exhibit all the desired characteristics of an effective drug delivery system for the treatment of pulmonary diseases that result primarily from ATII cell dysfunction.


Asunto(s)
Células Epiteliales Alveolares/inmunología , Técnicas de Transferencia de Gen , Inmunoconjugados/inmunología , Liposomas/inmunología , MicroARNs/administración & dosificación , Proteína C Asociada a Surfactante Pulmonar/inmunología , Animales , Línea Celular , Células Cultivadas , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Liposomas/química , Liposomas/farmacocinética , Pulmón/inmunología , Ratones Endogámicos C57BL
15.
Future Microbiol ; 9(10): 1151-63, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25405885

RESUMEN

IFITM3 restricts cellular infection by multiple important viral pathogens, and is particularly critical for the innate immune response against influenza virus. Expression of IFITM3 expands acidic endolysosomal compartments and prevents fusion of endocytosed viruses, leading to their degradation. This small, 133 amino acid, antiviral protein is controlled by at least four distinct post-translational modifications. Positive regulation of IFITM3 antiviral activity is provided by S-palmitoylation, while negative regulatory mechanisms include lysine ubiquitination, lysine methylation and tyrosine phosphorylation. Herein, we describe specific insights into IFITM3 trafficking and activity that were provided by studies of IFITM3 post-translational modifications, and discuss evidence suggesting that IFITM3 adopts multiple membrane topologies involving at least one intramembrane domain in its antivirally active conformation.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Orthomyxoviridae/inmunología , Procesamiento Proteico-Postraduccional , Proteínas de Unión al ARN/metabolismo , Membrana Celular/metabolismo , Modelos Biológicos , Modelos Moleculares , Conformación Proteica , Transporte de Proteínas
16.
J Phycol ; 50(1): 187-202, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26988018

RESUMEN

Twenty-six strains morphologically identified as Cylindrospermum as well as the closely related taxon Cronbergia siamensis were examined microscopically as well as phylogenetically using sequence data for the 16S rRNA gene and the 16S-23S internal transcribed spacer (ITS) region. Phylogenetic analysis of the 16S rRNA revealed three distinct clades. The clade we designate as Cylindrospermum sensu stricto contained all five of the foundational species, C. maius, C. stagnale, C. licheniforme, C. muscicola, and C. catenatum. In addition to these taxa, three species new to science in this clade were described: C. badium, C. moravicum, and C. pellucidum. Our evidence indicated that Cronbergia is a later synonym of Cylindrospermum. The phylogenetic position of Cylindrospermum within the Nostocaceae was not clearly resolved in our analyses. Cylindrospermum is unusual among cyanobacterial genera in that the morphological diversity appears to be more evident than sequence divergence. Taxa were clearly separable using morphology, but had very high percent similarity among ribosomal sequences. Given the high diversity we noted in this study, we conclude that there is likely much more diversity remaining to be described in this genus.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA