Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(37): 14906-11, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22927413

RESUMO

Yeast prions constitute a "protein-only" mechanism of inheritance that is widely deployed by wild yeast to create diverse phenotypes. One of the best-characterized prions, [PSI(+)], is governed by a conformational change in the prion domain of Sup35, a translation-termination factor. When this domain switches from its normal soluble form to an insoluble amyloid, the ensuing change in protein synthesis creates new traits. Two factors make these traits heritable: (i) the amyloid conformation is self-templating; and (ii) the protein-remodeling factor heat-shock protein (Hsp)104 (acting together with Hsp70 chaperones) partitions the template to daughter cells with high fidelity. Prions formed by several other yeast proteins create their own phenotypes but share the same mechanistic basis of inheritance. Except for the amyloid fibril itself, the cellular architecture underlying these protein-based elements of inheritance is unknown. To study the 3D arrangement of prion assemblies in their cellular context, we examined yeast [PSI(+)] prions in the native, hydrated state in situ, taking advantage of recently developed methods for cryosectioning of vitrified cells. Cryo-electron tomography of the vitrified sections revealed the prion assemblies as aligned bundles of regularly spaced fibrils in the cytoplasm with no bounding structures. Although the fibers were widely spaced, other cellular complexes, such as ribosomes, were excluded from the fibril arrays. Subtomogram image averaging, made possible by the organized nature of the assemblies, uncovered the presence of an additional array of densities between the fibers. We suggest these structures constitute a self-organizing mechanism that coordinates fiber deposition and the regulation of prion inheritance.


Assuntos
Padrões de Herança/genética , Modelos Moleculares , Príons/química , Conformação Proteica , Leveduras/genética , Microscopia Crioeletrônica , Processamento de Imagem Assistida por Computador , Microscopia de Fluorescência
2.
PLoS Pathog ; 8(12): e1003056, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23236278

RESUMO

All positive strand RNA viruses are known to replicate their genomes in close association with intracellular membranes. In case of the hepatitis C virus (HCV), a member of the family Flaviviridae, infected cells contain accumulations of vesicles forming a membranous web (MW) that is thought to be the site of viral RNA replication. However, little is known about the biogenesis and three-dimensional structure of the MW. In this study we used a combination of immunofluorescence- and electron microscopy (EM)-based methods to analyze the membranous structures induced by HCV in infected cells. We found that the MW is derived primarily from the endoplasmic reticulum (ER) and contains markers of rough ER as well as markers of early and late endosomes, COP vesicles, mitochondria and lipid droplets (LDs). The main constituents of the MW are single and double membrane vesicles (DMVs). The latter predominate and the kinetic of their appearance correlates with kinetics of viral RNA replication. DMVs are induced primarily by NS5A whereas NS4B induces single membrane vesicles arguing that MW formation requires the concerted action of several HCV replicase proteins. Three-dimensional reconstructions identify DMVs as protrusions from the ER membrane into the cytosol, frequently connected to the ER membrane via a neck-like structure. In addition, late in infection multi-membrane vesicles become evident, presumably as a result of a stress-induced reaction. Thus, the morphology of the membranous rearrangements induced in HCV-infected cells resemble those of the unrelated picorna-, corona- and arteriviruses, but are clearly distinct from those of the closely related flaviviruses. These results reveal unexpected similarities between HCV and distantly related positive-strand RNA viruses presumably reflecting similarities in cellular pathways exploited by these viruses to establish their membranous replication factories.


Assuntos
Retículo Endoplasmático , Hepacivirus , Hepatite C , Membranas Intracelulares , RNA Viral/biossíntese , Linhagem Celular , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Retículo Endoplasmático/virologia , Hepacivirus/fisiologia , Hepacivirus/ultraestrutura , Hepatite C/metabolismo , Hepatite C/patologia , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Membranas Intracelulares/virologia , Microscopia Eletrônica de Transmissão/métodos , Replicação Viral/fisiologia
3.
Blood ; 119(3): 786-97, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22123847

RESUMO

The Nef protein of HIV-1 facilitates viral replication and disease progression in vivo. Nef disturbs the organization of immunological synapses between infected CD4(+) T lymphocytes and antigen-presenting B-lymphocytes to interfere with TCR proximal signaling. Paradoxically, Nef enhances distal TCR signaling in infected CD4(+) T lymphocytes, an effect thought to be involved in its role in AIDS pathogenesis. Using quantitative confocal microscopy and cell fractionation of Nef-expressing cells and HIV-1-infected primary human T lymphocytes, we found that Nef induces intracellular compartmentalization of TCR signaling to adjust TCR responses to antigenic stimulation. Nef reroutes kinase-active pools of the TCR signaling master switch Lck away from the plasma membrane (PM) to the trans-Golgi network (TGN), thereby preventing the recruitment of active Lck to the immunological synapse after TCR engagement and limiting signal initiation at the PM. Instead, Nef triggers Lck-dependent activation of TGN-associated Ras-Erk signaling to promote the production of the T lymphocyte survival factor IL-2 and to enhance virus spread. Overexpression of the Lck PM transporter Unc119 restores Nef-induced subversions of Lck trafficking and TCR signaling. Nef therefore hijacks Lck sorting to selectively activate TGN-associated arms of compartmentalized TCR signaling. By tailoring T-lymphocyte responses to antigenic stimulation, Nef optimizes the environment for HIV-1 replication.


Assuntos
Linfócitos B/imunologia , Sinapses Imunológicas/fisiologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Rede trans-Golgi/imunologia , Linfócitos B/metabolismo , Linfócitos B/virologia , Comunicação Celular , Ensaio de Imunoadsorção Enzimática , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/metabolismo , Humanos , Interleucina-2/metabolismo , Ativação Linfocitária , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/imunologia , Receptores de Antígenos de Linfócitos T , Linfócitos T/metabolismo , Linfócitos T/virologia , Replicação Viral/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia , Rede trans-Golgi/metabolismo , Rede trans-Golgi/virologia
4.
Cell Microbiol ; 15(2): 292-304, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23279151

RESUMO

Human immunodeficiency virus type 1 (HIV-1) is a retrovirus that obtains its lipid envelope by budding through the plasma membrane of infected host cells. Various studies indicated that the HIV-1 membrane differs from the producer cell plasma membrane suggesting virus budding from pre-existing subdomains or virus-mediated induction of a specialized budding membrane. To perform a comparative lipidomics analysis by quantitative mass spectrometry, we first evaluated two independent methods to isolate the cellular plasma membrane. Subsequent lipid analysis of plasma membranes and HIV-1 purified from two different cell lines revealed a significantly different lipid composition of the viral membrane compared with the host cell plasma membrane, independent of the cell type investigated. Virus particles were significantly enriched in phosphatidylserine, sphingomyelin, hexosylceramide and saturated phosphatidylcholine species when compared with the host cell plasma membrane of the producer cells; they showed reduced levels of unsaturated phosphatidylcholine species, phosphatidylethanolamine and phosphatidylinositol. Cell type-specific differences in the lipid composition of HIV-1 and donor plasmamembranes were observed for plasmalogen-phosphatidylethanolamine and phosphatidylglycerol, which were strongly enriched only in HIV-1 derived from MT-4 cells. MT-4 cell-derived HIV-1 also contained dihydrosphingomyelin as reported previously, but this lipid class was also enriched in the host cell membrane. Taken together, these data strongly support the hypothesis that HIV-1 selects a specific lipid environment for its morphogenesis.


Assuntos
HIV-1/química , Microdomínios da Membrana/química , Vírion/química , Fracionamento Celular , Linhagem Celular , Ceramidas/análise , HIV-1/fisiologia , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Humanos , Espectrometria de Massas , Microdomínios da Membrana/fisiologia , Fosfatidilcolinas/análise , Fosfatidilinositóis/análise , Fosfatidilserinas/análise , Esfingomielinas/análise , Vírion/fisiologia
5.
PLoS Pathog ; 6(11): e1001173, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21124872

RESUMO

The structure of immature and mature HIV-1 particles has been analyzed in detail by cryo electron microscopy, while no such studies have been reported for cellular HIV-1 budding sites. Here, we established a system for studying HIV-1 virus-like particle assembly and release by cryo electron tomography of intact human cells. The lattice of the structural Gag protein in budding sites was indistinguishable from that of the released immature virion, suggesting that its organization is determined at the assembly site without major subsequent rearrangements. Besides the immature lattice, a previously not described Gag lattice was detected in some budding sites and released particles; this lattice was found at high frequencies in a subset of infected T-cells. It displays the same hexagonal symmetry and spacing in the MA-CA layer as the immature lattice, but lacks density corresponding to NC-RNA-p6. Buds and released particles carrying this lattice consistently lacked the viral ribonucleoprotein complex, suggesting that they correspond to aberrant products due to premature proteolytic activation. We hypothesize that cellular and/or viral factors normally control the onset of proteolytic maturation during assembly and release, and that this control has been lost in a subset of infected T-cells leading to formation of aberrant particles.


Assuntos
Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , HIV-1/química , HIV-1/ultraestrutura , RNA Viral/química , Vírion/química , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química , Células Cultivadas , Glioblastoma/metabolismo , HIV-1/fisiologia , Humanos , RNA Viral/metabolismo , Linfócitos T/virologia , Vírion/fisiologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
6.
Nat Med ; 11(7): 791-6, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15951748

RESUMO

Hepatitis C virus (HCV) infection causes chronic liver diseases and is a global public health problem. Detailed analyses of HCV have been hampered by the lack of viral culture systems. Subgenomic replicons of the JFH1 genotype 2a strain cloned from an individual with fulminant hepatitis replicate efficiently in cell culture. Here we show that the JFH1 genome replicates efficiently and supports secretion of viral particles after transfection into a human hepatoma cell line (Huh7). Particles have a density of about 1.15-1.17 g/ml and a spherical morphology with an average diameter of about 55 nm. Secreted virus is infectious for Huh7 cells and infectivity can be neutralized by CD81-specific antibodies and by immunoglobulins from chronically infected individuals. The cell culture-generated HCV is infectious for chimpanzee. This system provides a powerful tool for studying the viral life cycle and developing antiviral strategies.


Assuntos
Genoma Viral , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/patogenicidade , Animais , Anticorpos/farmacologia , Antígenos CD/imunologia , Fenômenos Biofísicos , Biofísica , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Clonagem Molecular , Hepacivirus/isolamento & purificação , Hepatite C/tratamento farmacológico , Hepatite C/imunologia , Humanos , Soros Imunes , Neoplasias Hepáticas/virologia , Microscopia Eletrônica , Pan troglodytes , RNA Viral , Tetraspanina 28 , Transfecção , Cultura de Vírus/métodos
7.
J Virol ; 84(9): 4646-58, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20147389

RESUMO

CD317/Bst-2/tetherin is a host factor that restricts the release of human immunodeficiency virus type 1 (HIV-1) by trapping virions at the plasma membrane of certain producer cells. It is antagonized by the HIV-1 accessory protein Vpu. Previous light microscopy studies localized CD317 to the plasma membrane and the endosomal compartment and showed Vpu induced downregulation. In the present study, we performed quantitative immunoelectron microscopy of CD317 in cells producing wild-type or Vpu-defective HIV-1 and in control cells. Double-labeling experiments revealed that CD317 localizes to the plasma membrane, to early and recycling endosomes, and to the trans-Golgi network. CD317 largely relocated to endosomes upon HIV-1 infection, and this effect was partly counteracted by Vpu. Unexpectedly, CD317 was enriched in the membrane of viral buds and cell-associated and cell-free viruses compared to the respective plasma membrane, and this enrichment was independent of Vpu. These results suggest that the tethering activity of CD317 critically depends on its density at the cell surface and appears to be less affected by its density in the virion membrane.


Assuntos
Antígenos CD/análise , Membrana Celular/química , Regulação da Expressão Gênica , HIV-1/química , Interações Hospedeiro-Patógeno , Glicoproteínas de Membrana/análise , Linhagem Celular , Endossomos/química , Proteínas Ligadas por GPI , Deleção de Genes , Proteínas do Vírus da Imunodeficiência Humana/deficiência , Proteínas do Vírus da Imunodeficiência Humana/fisiologia , Humanos , Microscopia Imunoeletrônica , Proteínas Virais Reguladoras e Acessórias/deficiência , Proteínas Virais Reguladoras e Acessórias/fisiologia , Rede trans-Golgi/química
8.
Nat Cell Biol ; 5(9): 793-802, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12942085

RESUMO

Pathogenic mycobacteria such as Mycobacterium tuberculosis and Mycobacterium avium facilitate disease by surviving intracellularly within a potentially hostile environment: the macrophage phagosome. They inhibit phagosome maturation processes, including fusion with lysosomes, acidification and, as shown here, membrane actin assembly. An in vitro assay developed for latex bead phagosomes (LBPs) provided insights into membrane signalling events that regulate phagosome actin assembly, a process linked to membrane fusion. Different lipids were found to stimulate or inhibit actin assembly by LBPs and mycobacterial phagosomes in vitro. In addition, selected lipids activated actin assembly and phagosome maturation in infected macrophages, resulting in a significant killing of M. tuberculosis and M. avium. In contrast, the polyunsaturated sigma-3 lipids behaved differently and stimulated pathogen growth. Thus, lipids can be involved in both stimulatory and inhibitory signalling networks in the phagosomal membrane.


Assuntos
Actinas/biossíntese , Metabolismo dos Lipídeos , Macrófagos/microbiologia , Mycobacteriaceae/metabolismo , Fagocitose/fisiologia , Fagossomos/microbiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Retroalimentação Fisiológica/efeitos dos fármacos , Retroalimentação Fisiológica/fisiologia , Interações Hospedeiro-Parasita/efeitos dos fármacos , Interações Hospedeiro-Parasita/fisiologia , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Lipídeos/farmacologia , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Fusão de Membrana/efeitos dos fármacos , Fusão de Membrana/fisiologia , Camundongos , Mycobacteriaceae/patogenicidade , Mycobacterium avium/efeitos dos fármacos , Mycobacterium avium/metabolismo , Mycobacterium avium/patogenicidade , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Óxido Nítrico/metabolismo , Fagocitose/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
9.
Nature ; 424(6949): 689-94, 2003 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-12894213

RESUMO

In metazoa, the nuclear envelope breaks down and reforms during each cell cycle. Nuclear pore complexes (NPCs), which serve as channels for transport between the nucleus and cytoplasm, assemble into the reforming nuclear envelope in a sequential process involving association of a subset of NPC proteins, nucleoporins, with chromatin followed by the formation of a closed nuclear envelope fenestrated by NPCs. How chromatin recruitment of nucleoporins and NPC assembly are regulated is unknown. Here we demonstrate that RanGTP production is required to dissociate nucleoporins Nup107, Nup153 and Nup358 from Importin beta, to target them to chromatin and to induce association between separate NPC subcomplexes. Additionally, either an excess of RanGTP or removal of Importin beta induces formation of NPC-containing membrane structures--annulate lamellae--both in vitro in the absence of chromatin and in vivo. Annulate lamellae formation is strongly and specifically inhibited by an excess of Importin beta. The data demonstrate that RanGTP triggers distinct steps of NPC assembly, and suggest a mechanism for the spatial restriction of NPC assembly to the surface of chromatin.


Assuntos
Cromatina/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Poro Nuclear/química , Poro Nuclear/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Substituição de Aminoácidos , Animais , Extratos Celulares , Feminino , Masculino , Mutação , Oócitos , Fosforilação , Transporte Proteico , Interferência de RNA , Espermatozoides , Xenopus laevis , beta Carioferinas/metabolismo , Proteína ran de Ligação ao GTP/genética
10.
PLoS Pathog ; 3(3): e36, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17381240

RESUMO

HIV-1 assembly and release are believed to occur at the plasma membrane in most host cells with the exception of primary macrophages, for which exclusive budding at late endosomes has been reported. Here, we applied a novel ultrastructural approach to assess HIV-1 budding in primary macrophages in an immunomarker-independent manner. Infected macrophages were fed with BSA-gold and stained with the membrane-impermeant dye ruthenium red to identify endosomes and the plasma membrane, respectively. Virus-filled vacuolar structures with a seemingly intracellular localization displayed intense staining with ruthenium red, but lacked endocytosed BSA-gold, defining them as plasma membrane. Moreover, HIV budding profiles were virtually excluded from gold-filled endosomes while frequently being detected on ruthenium red-positive membranes. The composition of cellular marker proteins incorporated into HIV-1 supported a plasma membrane-derived origin of the viral envelope. Thus, contrary to current opinion, the plasma membrane is the primary site of HIV-1 budding also in infected macrophages.


Assuntos
Membrana Celular/virologia , HIV-1/crescimento & desenvolvimento , Macrófagos/virologia , Morfogênese/fisiologia , Antígenos CD/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Células Cultivadas , Endossomos/ultraestrutura , Ouro , HIV-1/fisiologia , Humanos , Proteína Kangai-1/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Dados de Sequência Molecular , Glicoproteínas da Membrana de Plaquetas/metabolismo , Rutênio Vermelho , Tetraspanina 28 , Tetraspanina 30 , Vacúolos/ultraestrutura , Vacúolos/virologia
11.
Mol Biol Cell ; 15(1): 345-58, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14617814

RESUMO

Actin is implicated in membrane fusion, but the precise mechanisms remain unclear. We showed earlier that membrane organelles catalyze the de novo assembly of F-actin that then facilitates the fusion between latex bead phagosomes and a mixture of early and late endocytic organelles. Here, we correlated the polymerization and organization of F-actin with phagosome and endocytic organelle fusion processes in vitro by using biochemistry and light and electron microscopy. When membrane organelles and cytosol were incubated at 37 degrees C with ATP, cytosolic actin polymerized rapidly and became organized into bundles and networks adjacent to membrane organelles. By 30-min incubation, a gel-like state was formed with little further polymerization of actin thereafter. Also during this time, the bulk of in vitro fusion events occurred between phagosomes/endocytic organelles. The fusion between latex bead phagosomes and late endocytic organelles, or between late endocytic organelles themselves was facilitated by actin, but we failed to detect any effect of perturbing F-actin polymerization on early endosome fusion. Consistent with this, late endosomes, like phagosomes, could nucleate F-actin, whereas early endosomes could not. We propose that actin assembled by phagosomes or late endocytic organelles can provide tracks for fusion-partner organelles to move vectorially toward them, via membrane-bound myosins, to facilitate fusion.


Assuntos
Actinas/metabolismo , Endossomos/metabolismo , Macrófagos/metabolismo , Fusão de Membrana/fisiologia , Fagossomos/metabolismo , Trifosfato de Adenosina , Animais , Células Cultivadas , Microscopia Crioeletrônica , Citoesqueleto/metabolismo , Citosol/metabolismo , Endocitose/fisiologia , Camundongos , Microscopia Confocal , Modelos Moleculares , Organelas/metabolismo , Timosina/metabolismo
12.
JCI Insight ; 1(9)2016 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-27430022

RESUMO

Vertebrate life critically depends on renal filtration and excretion of low molecular weight waste products. This process is controlled by a specialized cell-cell contact between podocyte foot processes: the slit diaphragm (SD). Using a comprehensive set of targeted KO mice of key SD molecules, we provided genetic, functional, and high-resolution ultrastructural data highlighting a concept of a flexible, dynamic, and multilayered architecture of the SD. Our data indicate that the mammalian SD is composed of NEPHRIN and NEPH1 molecules, while NEPH2 and NEPH3 do not participate in podocyte intercellular junction formation. Unexpectedly, homo- and heteromeric NEPHRIN/NEPH1 complexes are rarely observed. Instead, single NEPH1 molecules appear to form the lower part of the junction close to the glomerular basement membrane with a width of 23 nm, while single NEPHRIN molecules form an adjacent junction more apically with a width of 45 nm. In both cases, the molecules are quasiperiodically spaced 7 nm apart. These structural findings, in combination with the flexibility inherent to the repetitive Ig folds of NEPHRIN and NEPH1, indicate that the SD likely represents a highly dynamic cell-cell contact that forms an adjustable, nonclogging barrier within the renal filtration apparatus.

13.
Cell Host Microbe ; 17(1): 58-71, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25500191

RESUMO

The host endolysosomal compartment is often manipulated by intracellular bacterial pathogens. Salmonella (Salmonella enterica serovar Typhimurium) secrete numerous effector proteins, including SifA, through a specialized type III secretion system to hijack the host endosomal system and generate the Salmonella-containing vacuole (SCV). To form this replicative niche, Salmonella targets the Rab7 GTPase to recruit host membranes through largely unknown mechanisms. We show that Pleckstrin homology domain-containing protein family member 1 (PLEKHM1), a lysosomal adaptor, is targeted by Salmonella through direct interaction with SifA. By binding the PLEKHM1 PH2 domain, Salmonella utilize a complex containing PLEKHM1, Rab7, and the HOPS tethering complex to mobilize phagolysosomal membranes to the SCV. Depletion of PLEKHM1 causes a profound defect in SCV morphology with multiple bacteria accumulating in enlarged structures and significantly dampens Salmonella proliferation in multiple cell types and mice. Thus, PLEKHM1 provides a critical interface between pathogenic infection and the host endolysosomal system.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Bactérias/metabolismo , Glicoproteínas/metabolismo , Interações Hospedeiro-Patógeno , Glicoproteínas de Membrana/metabolismo , Salmonella typhimurium/crescimento & desenvolvimento , Vacúolos/microbiologia , Animais , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
14.
J Histochem Cytochem ; 52(8): 991-1000, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15258174

RESUMO

Particulate gold labeling on ultrathin sections is in widespread use for antigen localization at the EM level. To extend the usefulness of gold labeling technology, we are evaluating different methods for sampling and estimating quantities of gold labeling. Here we present a simple, rapid, and unbiased method for assessing the relative pool sizes of immunogold labeling distributed over different cell compartments. The method uses a sampling approach developed for stereology in which a regular array of microscopic fields or linear scans is positioned randomly on labeled sections. From these readouts, gold particles are counted and assigned to identifiable cell structures to construct a gold labeling frequency distribution of those labeled compartments. Here we use ultrathin cryosections labeled for a range of different proteins and for a signaling lipid. We show by scanning labeled sections at the electron microscope that counting 100-200 particles on each of two grids is sufficient to obtain a reproducible and rapid assessment of the pattern of labeling proportions over 10-16 compartments. If more precise estimates of labeling proportions over individual compartments are required (e.g., to achieve coefficients of error of 10-20%), then 100-200 particles need to be counted over each compartment of interest.


Assuntos
Imuno-Histoquímica/métodos , Coloração e Rotulagem/métodos , Animais , Linhagem Celular , Camundongos , Microscopia Eletrônica , Microtomia , Reprodutibilidade dos Testes
15.
Cell Host Microbe ; 5(3): 285-97, 2009 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-19286137

RESUMO

Mammals encode proteins that inhibit viral replication at the cellular level. In turn, certain viruses have evolved genes that can functionally counteract these intrinsic restrictions. Human CD317 (BST-2/HM1.24/tetherin) is a restriction factor that blocks release of human immunodeficiency virus type 1 (HIV-1) from the cell surface and can be overcome by HIV-1 Vpu. Here, we show that mouse and rat CD317 potently inhibit HIV-1 release but are resistant to Vpu. Interspecies chimeras reveal that the rodent-specific resistance and human-specific sensitivity to Vpu antagonism involve all three major structural domains of CD317. To promote virus release, Vpu depletes cellular pools of human CD317, but not of the rodent orthologs, by accelerating its degradation via the 20S proteasome. Thus, HIV-1 Vpu suppresses the expression of the CD317 antiviral factor in human cells, and the species-specific resistance to this suppression may guide the development of small animal models of HIV infection.


Assuntos
Antígenos CD/imunologia , HIV-1/imunologia , HIV-1/patogenicidade , Proteínas do Vírus da Imunodeficiência Humana/fisiologia , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/imunologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Fatores de Virulência/fisiologia , Animais , Antígenos CD/metabolismo , Linhagem Celular , Proteínas Ligadas por GPI , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos , Ratos
16.
J Biol Chem ; 283(46): 32024-33, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18772135

RESUMO

Morphogenesis of infectious HIV-1 involves budding of immature virions followed by proteolytic disassembly of the Gag protein shell and subsequent assembly of processed capsid proteins (CA) into the mature HIV-1 core. The dimeric interface between C-terminal domains of CA (C-CA) has been shown to be important for both immature and mature assemblies. We previously reported a CA-binding peptide (CAI) that blocks both assembly steps in vitro. The three-dimensional structure of the C-CA/CAI complex revealed an allosteric effect of CAI that alters the C-CA dimer interface. Based on this structure, we now investigated the phenotypes of mutations in the binding pocket. CA variants carrying mutations Y169A, L211A, or L211S had a reduced affinity for CAI and were unable to form mature-like particles in vitro. These mutations also blocked morphological conversion to mature virions in tissue culture and abolished infectivity. X-ray crystallographic analyses of the variant C-CA domains revealed that these alterations induced the same allosteric change at the dimer interface observed in the C-CA/CAI complex. These results point to a role of key interactions between conserved amino acids in the CAI binding pocket of C-CA in maintaining the correct conformation necessary for mature core assembly.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Capsídeo/efeitos dos fármacos , Capsídeo/metabolismo , HIV-1/química , HIV-1/metabolismo , Montagem de Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Sítios de Ligação , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/ultraestrutura , Linhagem Celular , Sequência Conservada , Cristalografia por Raios X , HIV-1/efeitos dos fármacos , HIV-1/ultraestrutura , Humanos , Microscopia Eletrônica , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Peptídeos/farmacologia , Estrutura Quaternária de Proteína , Alinhamento de Sequência
17.
J Cell Biol ; 182(4): 685-701, 2008 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-18725538

RESUMO

Autophagy is the engulfment of cytosol and organelles by double-membrane vesicles termed autophagosomes. Autophagosome formation is known to require phosphatidylinositol 3-phosphate (PI(3)P) and occurs near the endoplasmic reticulum (ER), but the exact mechanisms are unknown. We show that double FYVE domain-containing protein 1, a PI(3)P-binding protein with unusual localization on ER and Golgi membranes, translocates in response to amino acid starvation to a punctate compartment partially colocalized with autophagosomal proteins. Translocation is dependent on Vps34 and beclin function. Other PI(3)P-binding probes targeted to the ER show the same starvation-induced translocation that is dependent on PI(3)P formation and recognition. Live imaging experiments show that this punctate compartment forms near Vps34-containing vesicles, is in dynamic equilibrium with the ER, and provides a membrane platform for accumulation of autophagosomal proteins, expansion of autophagosomal membranes, and emergence of fully formed autophagosomes. This PI(3)P-enriched compartment may be involved in autophagosome biogenesis. Its dynamic relationship with the ER is consistent with the idea that the ER may provide important components for autophagosome formation.


Assuntos
Autofagia , Compartimento Celular , Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Fagossomos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Sequência de Aminoácidos , Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Linhagem Celular , Sobrevivência Celular , Células Clonais , Regulação para Baixo , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/ultraestrutura , Proteínas de Fluorescência Verde/metabolismo , Humanos , Membranas Intracelulares/ultraestrutura , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Fagossomos/enzimologia , Fagossomos/ultraestrutura , Fosfatidilinositol 3-Quinases/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico , Ratos , Proteínas Recombinantes de Fusão/metabolismo
18.
J Virol ; 80(13): 6267-75, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16775314

RESUMO

The p6 domain of human immunodeficiency virus type 1 (HIV-1) Gag has long been known to be monoubiquitinated. We have previously shown that the MA, CA, and NC domains are also monoubiquitinated at low levels (E. Gottwein and H. G. Krausslich, J. Virol. 79:9134-9144, 2005). While several lines of evidence support a role for ubiquitin in virus release, the relevance of Gag ubiquitination is unclear. To directly address the function of Gag ubiquitination, we constructed Gag variants in which lysine residues in the NC, SP2, and p6 domains were mutated to arginine either in individual domains or in combination. Using these mutants, we showed that in addition to MA, CA, NC, and p6, SP2 is also mono- or di-ubiquitinated at levels comparable to those of the other domains. Replacement of all lysine residues in only one of the domains had minor effects on virus release, while cumulative mutations in NC and SP2 or in NC and p6 resulted in an accumulation of late budding structures, as observed by electron microscopy analysis. Strikingly, replacement of all lysine residues downstream of CA led to a significant reduction in virus release kinetics and a fivefold accumulation of late viral budding structures compared to wild-type levels. These results indicate that ubiquitination of lysine residues in Gag in the vicinity of the viral late domain is important for HIV-1 budding, while no specific lysine residue may be needed and individual domains can functionally substitute. This is consistent with Gag ubiquitination being functionally involved in a transient protein interaction network at the virus budding site.


Assuntos
Produtos do Gene gag/metabolismo , HIV-1/metabolismo , Mutação , Processamento de Proteína Pós-Traducional , Ubiquitina/metabolismo , Eliminação de Partículas Virais , Substituição de Aminoácidos , Arginina/genética , Arginina/metabolismo , Produtos do Gene gag/genética , HIV-1/genética , HIV-1/ultraestrutura , Células HeLa , Humanos , Lisina/genética , Lisina/metabolismo , Microscopia Eletrônica de Transmissão , Processamento de Proteína Pós-Traducional/genética , Estrutura Terciária de Proteína/genética , Eliminação de Partículas Virais/genética
19.
Traffic ; 7(11): 1551-66, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17014699

RESUMO

The endosomal sorting complex required for transport (ESCRT) is thought to support the formation of intralumenal vesicles of multivesicular bodies (MVBs). The ESCRT is also required for the budding of HIV and has been proposed to be recruited to the HIV-budding site, the plasma membrane of T cells and MVBs in macrophages. Despite increasing data on the function of ESCRT, the ultrastructural localization of its components has not been determined. We therefore localized four proteins of the ESCRT machinery in human T cells and macrophages by quantitative electron microscopy. All the proteins were found throughout the endocytic pathway, including the plasma membrane, with only around 10 and 3% of the total labeling in the cytoplasm and on the MVBs, respectively. The majority of the labeling (45%) was unexpectedly found on tubular-vesicular endosomal membranes rather than on endosomes themselves. The ESCRT labeling was twice as concentrated on early and late endosomes/lysosomes in macrophages compared with that in T cells, where it was twice more abundant at the plasma membrane. The ESCRT proteins were not redistributed on HIV infection, suggesting that the amount of ESCRT proteins located at the budding site suffices for HIV release. These results represent the first systematic ultrastructural localization of ESCRT and provide insights into its role in uninfected and HIV-infected cells.


Assuntos
Endossomos/metabolismo , Membranas Intracelulares/metabolismo , Vesículas Transportadoras/metabolismo , Proteínas de Transporte Vesicular/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/ultraestrutura , Linfócitos T CD4-Positivos/virologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Citosol/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte , Endossomos/ultraestrutura , HIV-1/crescimento & desenvolvimento , HIV-1/metabolismo , Células HeLa , Humanos , Membranas Intracelulares/ultraestrutura , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Macrófagos/virologia , Microscopia Eletrônica , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Linfócitos T/metabolismo , Linfócitos T/ultraestrutura , Linfócitos T/virologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Vesículas Transportadoras/ultraestrutura , Proteínas de Transporte Vesicular/genética , Vírion/metabolismo
20.
Histochem Cell Biol ; 119(4): 333-41, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12695916

RESUMO

Quantitative immunoelectron microscopy of gold label in intracellular compartments often involves calculating labelling densities (LDs). These are related to antigen concentrations and usually refer gold particle counts to the sizes of compartments on sections (for example, golds per microm(2) of organelle profile area or per microm of membrane trace length). Here, we show how LD values can be estimated more simply (without estimating areas or lengths) and also how observed and expected LD values can be used to calculate a relative labelling index (RLI) for each compartment and then test statistically for preferential (non-random) labelling. For random labelling, RLI=1. Compartment size is estimated stereologically by superimposing random test points (which hit organelle profiles in proportion to their area) or test lines (which intersect membrane traces in proportion to their length). By this means, the observed LD of a compartment (LD(obs)) can be expressed simply as golds per test point (organelles) or per intersection (membranes). Furthermore, the LD obtained by dividing total golds (on all compartments) by total points or intersections (on all compartments) is the value to be expected (LD(exp)) when compartments label randomly. For each compartment, RLI=LD(obs)/LD(exp). Statistical analysis is undertaken by comparing observed distributions of golds with predicted random distributions (calculated from point or intersection counts). A compartment is preferentially labelled if two criteria are met: (1) its RLI>1 (i.e. LD(obs) is greater than LD(exp)) and (2) its partial chi-squared value makes a substantial contribution to total chi-squared value. This approach provides a simple and efficient way of comparing LDs in different compartments. Its utility is illustrated using data from VPARP and LAMP-1 labelling experiments.


Assuntos
Antígenos CD/análise , Imuno-Histoquímica , Poli(ADP-Ribose) Polimerases/análise , Coloração e Rotulagem/métodos , Animais , Células HeLa/química , Células HeLa/ultraestrutura , Humanos , Proteínas de Membrana Lisossomal , Macrófagos/química , Macrófagos/ultraestrutura , Camundongos , Microscopia Imunoeletrônica , Fagossomos/química , Fagossomos/ultraestrutura , Distribuição Aleatória , Partículas de Ribonucleoproteínas em Forma de Abóbada
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA