Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Pharmacol Drug Dev ; 12(2): 202-211, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36065092

RESUMEN

ZYIL1 is a nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome inhibitor, which prevents NLRP3-induced apoptosis-associated speck-like protein containing a caspase activation and recruitment domain oligomerization, thus inhibiting NLRP3 inflammasome pathway. We investigated the safety, tolerability, pharmacokinetic, and pharmacodynamic profiles of ZYIL1 after single and multiple doses in healthy subjects. The subjects aged 18-55 years were enrolled in 2 different studies: single and multiple ascending dose. Blood/urine samples were collected at designated time points for pharmacokinetic and pharmacodynamic analysis. In the single-ascending-dose study, 30 subjects were enrolled (6 subjects each in 5 dose groups). One adverse event was reported during the study. ZYIL1 was well absorbed with median time to maximum plasma concentration at 1-1.5 hours. The exposures were dose proportional across the dose ranges. ZYIL1 is excreted as an unchanged form via the renal route. The mean elimination half-life was 6-7 hours. In the multiple-ascending-dose study, 18 subjects were enrolled (6 subjects each in 3 dose groups). Eleven adverse events were reported by 6 subjects during the study. The accumulation index at steady state for area under the plasma concentration-time curve indicated that ZYIL1 has a marginal accumulation upon repeated dosing. Dose-proportional exposure was observed across the dose ranges. All subjects showed >90% interleukin (IL)-1ß inhibition in all dose groups for both studies. Inhibition in IL-1ß and IL-18 was observed throughout the 14 days of treatment in the multiple-dose study. The safety profile, rapid absorption, marginal accumulation, and significant inhibition of IL-1ß and IL-18 level support its development for the management of inflammatory disorders.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Interleucina-18/metabolismo , Área Bajo la Curva
2.
Bioorg Med Chem Lett ; 30(21): 127571, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32980515

RESUMEN

NLRP3 inflammasome mediated release of interleukin-1ß (IL-1ß) has been implicated in various diseases, including COVID-19. In this study, rationally designed alkenyl sulfonylurea derivatives were identified as novel, potent and orally bioavailable NLRP3 inhibitors. Compound 7 was found to be potent (IL-1ß IC50 = 35 nM; IL-18 IC50 = 33 nM) and selective NLRP3 inflammasome inhibitor with excellent pharmacokinetic profile having oral bioavailability of 99% in mice.


Asunto(s)
Inflamasomas/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Compuestos de Sulfonilurea/farmacología , Administración Oral , Animales , Betacoronavirus , COVID-19 , Línea Celular Tumoral , Infecciones por Coronavirus , Inhibidores del Citocromo P-450 CYP2C8/administración & dosificación , Inhibidores del Citocromo P-450 CYP2C8/síntesis química , Inhibidores del Citocromo P-450 CYP2C8/farmacocinética , Inhibidores del Citocromo P-450 CYP2C8/farmacología , Inhibidores del Citocromo P-450 CYP2C9/administración & dosificación , Inhibidores del Citocromo P-450 CYP2C9/síntesis química , Inhibidores del Citocromo P-450 CYP2C9/farmacocinética , Inhibidores del Citocromo P-450 CYP2C9/farmacología , Perros , Estabilidad de Medicamentos , Humanos , Interleucina-1beta/antagonistas & inhibidores , Ratones Endogámicos C57BL , Microsomas Hepáticos/metabolismo , Estructura Molecular , Pandemias , Neumonía Viral , Ratas , SARS-CoV-2 , Relación Estructura-Actividad , Compuestos de Sulfonilurea/administración & dosificación , Compuestos de Sulfonilurea/síntesis química , Compuestos de Sulfonilurea/farmacocinética
3.
ACS Med Chem Lett ; 11(4): 414-418, 2020 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-32292543

RESUMEN

NLRP3 inflammasome mediated release of interleukin-1ß (IL-1ß) has been implicated in various diseases. In this study, rationally designed mimics of sulfonylurea moiety were investigated as NLRP3 inhibitors. Our results culminated into discovery of series of unprecedented N-cyano sulfoximineurea derivatives as potent NLRP3 inflammasome inhibitors. Compound 15 (IC50 = 7 nM) and analogues were found to be highly potent and selective NLRP3 inflammasome inhibitor with good pharmacokinetic profile. These effects translate in vivo, as 15, 29, and 34 significantly inhibit NLRP3 dependent IL-1ß secretion in mice.

4.
PLoS One ; 12(11): e0187899, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29121670

RESUMEN

Human cytomegalovirus (HCMV) depends on and modulates multiple host cell membrane proteins during each stage of the viral life cycle. To gain a global view of the impact of HCMV-infection on membrane proteins, we analyzed HCMV-induced changes in the abundance of membrane proteins in fibroblasts using stable isotope labeling with amino acids (SILAC), membrane fractionation and protein identification by two-dimensional liquid chromatography and tandem mass spectrometry. This systematic approach revealed that CD81, CD44, CD98, caveolin-1 and catenin delta-1 were down-regulated during infection whereas GRP-78 was up-regulated. Since CD81 downregulation was also observed during infection with UV-inactivated virus we hypothesized that this tetraspanin is part of the viral entry process. Interestingly, additional members of the tetraspanin family, CD9 and CD151, were also downregulated during HCMV-entry. Since tetraspanin-enriched microdomains (TEM) cluster host cell membrane proteins including known CMV receptors such as integrins, we studied whether TEMs are required for viral entry. When TEMs were disrupted with the cholesterol chelator methyl-ß-cylcodextrin, viral entry was inhibited and this inhibition correlated with reduced surface levels of CD81, CD9 and CD151, whereas integrin levels remained unchanged. Furthermore, simultaneous siRNA-mediated knockdown of multiple tetraspanins inhibited viral entry whereas individual knockdown had little effect suggesting essential, but redundant roles for individual tetraspanins during entry. Taken together, our data suggest that TEM act as platforms for receptors utilized by HCMV for entry into cells.


Asunto(s)
Membrana Celular/metabolismo , Citomegalovirus/fisiología , Fibroblastos/virología , Proteómica/métodos , Tetraspaninas/metabolismo , Internalización del Virus , Línea Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Integrinas/química , Integrinas/metabolismo , Marcaje Isotópico , Dominios Proteicos/efectos de los fármacos , Tetraspaninas/química , Tetraspaninas/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , beta-Ciclodextrinas/farmacología
5.
J Clin Invest ; 124(5): 1928-44, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24691437

RESUMEN

The most abundantly produced virion protein in human cytomegalovirus (HCMV) is the immunodominant phosphoprotein 65 (pp65), which is frequently included in CMV vaccines. Although it is nonessential for in vitro CMV growth, pp65 displays immunomodulatory functions that support a potential role in primary and/or persistent infection. To determine the contribution of pp65 to CMV infection and immunity, we generated a rhesus CMV lacking both pp65 orthologs (RhCMVΔpp65ab). While deletion of pp65ab slightly reduced growth in vitro and increased defective particle formation, the protein composition of secreted virions was largely unchanged. Interestingly, pp65 was not required for primary and persistent infection in animals. Immune responses induced by RhCMVΔpp65ab did not prevent reinfection with rhesus CMV; however, reinfection with RhCMVΔUS2-11, which lacks viral-encoded MHC-I antigen presentation inhibitors, was prevented. Unexpectedly, induction of pp65b-specific T cells alone did not protect against RhCMVΔUS2-11 challenge, suggesting that T cells targeting multiple CMV antigens are required for protection. However, pp65-specific immunity was crucial for controlling viral dissemination during primary infection, as indicated by the marked increase of RhCMVΔpp65ab genome copies in CMV-naive, but not CMV-immune, animals. Our data provide rationale for inclusion of pp65 into CMV vaccines but also demonstrate that pp65-induced T cell responses alone do not recapitulate the protective effect of natural infection.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Fosfoproteínas/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Presentación de Antígeno/inmunología , Línea Celular , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/patología , Vacunas contra Citomegalovirus/genética , Vacunas contra Citomegalovirus/inmunología , Eliminación de Gen , Humanos , Macaca mulatta , Ratones , Fosfoproteínas/genética , Linfocitos T/inmunología , Linfocitos T/patología , Proteínas de la Matriz Viral/genética
6.
PLoS One ; 7(9): e44694, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23049756

RESUMEN

Poxviruses express highly active inhibitors, including serine proteinase inhibitors (serpins), designed to target host immune defense pathways. Recent work has demonstrated clinical efficacy for a secreted, myxomaviral serpin, Serp-1, which targets the thrombotic and thrombolytic proteases, suggesting that other viral serpins may have therapeutic application. Serp-2 and CrmA are intracellular cross-class poxviral serpins, with entirely distinct functions from the Serp-1 protein. Serp-2 and CrmA block the serine protease granzyme B (GzmB) and cysteine proteases, caspases 1 and 8, in apoptotic pathways, but have not been examined for extracellular anti-inflammatory activity. We examined the ability of these cross-class serpins to inhibit plaque growth after arterial damage or transplant and to reduce leukocyte apoptosis. We observed that purified Serp-2, but not CrmA, given as a systemic infusion after angioplasty, transplant, or cuff-compression injury markedly reduced plaque growth in mouse and rat models in vivo. Plaque growth was inhibited both locally at sites of surgical trauma, angioplasty or transplant, and systemically at non-injured sites in ApoE-deficient hyperlipidemic mice. With analysis in vitro of human cells in culture, Serp-2 selectively inhibited T cell caspase activity and blocked cytotoxic T cell (CTL) mediated killing of T lymphocytes (termed fratricide). Conversely, both Serp-2 and CrmA inhibited monocyte apoptosis. Serp-2 inhibitory activity was significantly compromised either in vitro with GzmB antibody or in vivo in ApoE/GzmB double knockout mice. Conclusions The viral cross-class serpin, Serp-2, that targets both apoptotic and inflammatory pathways, reduces vascular inflammation in a GzmB-dependent fashion in vivo, and inhibits human T cell apoptosis in vitro. These findings indicate that therapies targeting Granzyme B and/or T cell apoptosis may be used to inhibit T lymphocyte apoptosis and inflammation in response to arterial injury.


Asunto(s)
Aorta/efectos de los fármacos , Estenosis Carotídea/tratamiento farmacológico , Citotoxicidad Inmunológica/efectos de los fármacos , Inflamación/tratamiento farmacológico , Serpinas/farmacología , Linfocitos T/efectos de los fármacos , Proteínas Virales/farmacología , Angioplastia/efectos adversos , Animales , Aorta/inmunología , Aorta/trasplante , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Estenosis Carotídea/etiología , Estenosis Carotídea/inmunología , Estenosis Carotídea/patología , Caspasa 1/metabolismo , Caspasa 8/metabolismo , Línea Celular , Expresión Génica/efectos de los fármacos , Granzimas/antagonistas & inhibidores , Granzimas/metabolismo , Humanos , Inflamación/etiología , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Serpinas/genética , Serpinas/aislamiento & purificación , Linfocitos T/inmunología , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación
7.
J Virol ; 86(17): 8959-73, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22718821

RESUMEN

Cytomegaloviruses are highly host restricted, resulting in cospeciation with their hosts. As a natural pathogen of rhesus macaques (RM), rhesus cytomegalovirus (RhCMV) has therefore emerged as a highly relevant experimental model for pathogenesis and vaccine development due to its close evolutionary relationship to human CMV (HCMV). Most in vivo experiments performed with RhCMV employed strain 68-1 cloned as a bacterial artificial chromosome (BAC). However, the complete genome sequence of the 68-1 BAC has not been determined. Furthermore, the gene content of the RhCMV genome is unknown, and previous open reading frame (ORF) predictions relied solely on uninterrupted ORFs with an arbitrary cutoff of 300 bp. To obtain a more precise picture of the actual proteins encoded by the most commonly used molecular clone of RhCMV, we reevaluated the RhCMV 68-1 BAC genome by whole-genome shotgun sequencing and determined the protein content of the resulting RhCMV virions by proteomics. By comparing the RhCMV genome to those of several related Old World monkey (OWM) CMVs, we were able to filter out many unlikely ORFs and obtain a simplified map of the RhCMV genome. This comparative genomics analysis suggests a high degree of ORF conservation among OWM CMVs, thus decreasing the likelihood that ORFs found only in RhCMV comprise true genes. Moreover, virion proteomics independently validated the revised ORF predictions, since only proteins that were conserved across OWM CMVs could be detected. Taken together, these data suggest a much higher conservation of genome and virion structure between CMVs of humans, apes, and OWMs than previously assumed.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Infecciones por Citomegalovirus/veterinaria , Infecciones por Citomegalovirus/virología , Citomegalovirus/genética , Sistemas de Lectura Abierta , Enfermedades de los Primates/virología , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Cercopithecidae , Cromosomas Artificiales Bacterianos/metabolismo , Citomegalovirus/química , Citomegalovirus/clasificación , Citomegalovirus/metabolismo , Evolución Molecular , Genoma Viral , Humanos , Macaca mulatta , Datos de Secuencia Molecular , Filogenia , Proteómica , Alineación de Secuencia , Proteínas Virales/química
8.
PLoS Pathog ; 7(11): e1002332, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22072961

RESUMEN

Interferon-induced BST2/Tetherin prevents budding of vpu-deficient HIV-1 by tethering mature viral particles to the plasma membrane. BST2 also inhibits release of other enveloped viruses including Ebola virus and Kaposi's sarcoma associated herpesvirus (KSHV), indicating that BST2 is a broadly acting antiviral host protein. Unexpectedly however, recovery of human cytomegalovirus (HCMV) from supernatants of BST2-expressing human fibroblasts was increased rather than decreased. Furthermore, BST2 seemed to enhance viral entry into cells since more virion proteins were released into BST2-expressing cells and subsequent viral gene expression was elevated. A significant increase in viral entry was also observed upon induction of endogenous BST2 during differentiation of the pro-monocytic cell line THP-1. Moreover, treatment of primary human monocytes with siRNA to BST2 reduced HCMV infection, suggesting that BST2 facilitates entry of HCMV into cells expressing high levels of BST2 either constitutively or in response to exogenous stimuli. Since BST2 is present in HCMV particles we propose that HCMV entry is enhanced via a reverse-tethering mechanism with BST2 in the viral envelope interacting with BST2 in the target cell membrane. Our data suggest that HCMV not only counteracts the well-established function of BST2 as inhibitor of viral egress but also employs this anti-viral protein to gain entry into BST2-expressing hematopoietic cells, a process that might play a role in hematogenous dissemination of HCMV.


Asunto(s)
Antígenos CD/metabolismo , Citomegalovirus/fisiología , Internalización del Virus , Liberación del Virus , Antígenos CD/genética , Línea Celular , Ebolavirus/fisiología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regulación Viral de la Expresión Génica , Células HEK293 , VIH-1/fisiología , Herpesvirus Humano 8/fisiología , Humanos , Monocitos/virología , Interferencia de ARN , ARN Interferente Pequeño
9.
Mol Biol Cell ; 22(17): 3218-30, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21757542

RESUMEN

Following endocytosis, internalized plasma membrane proteins can be recycled back to the cell surface or trafficked to late endosomes/lysosomes for degradation. Here we report on the trafficking of multiple proteins that enter cells by clathrin-independent endocytosis (CIE) and determine that a set of proteins (CD44, CD98, and CD147) found primarily in recycling tubules largely failed to reach late endosomes in HeLa cells, whereas other CIE cargo proteins, including major histocompatibility complex class I protein (MHCI), trafficked to both early endosome antigen 1 (EEA1) and late endosomal compartments in addition to recycling tubules. Expression of the membrane-associated RING-CH 8 (MARCH8) E3 ubiquitin ligase completely shifted the trafficking of CD44 and CD98 proteins away from recycling tubules to EEA1 compartments and late endosomes, resulting in reduced surface levels. Cargo affected by MARCH expression, including CD44, CD98, and MHCI, still entered cells by CIE, suggesting that the routing of ubiquitinated cargo occurs after endocytosis. MARCH8 expression led to direct ubiquitination of CD98 and routing of CD98 to late endosomes/lysosomes.


Asunto(s)
Clatrina/metabolismo , Endosomas/metabolismo , Transporte de Proteínas , Ubiquitina-Proteína Ligasas/metabolismo , Basigina/metabolismo , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Proteínas de Unión al ADN/metabolismo , Endocitosis , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteína-1 Reguladora de Fusión/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Células HeLa , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Proteolisis , Factores de Transcripción/metabolismo , Ubiquitinación , Proteínas de Transporte Vesicular/metabolismo
10.
Photochem Photobiol ; 87(5): 1164-72, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21699546

RESUMEN

Erosion and rupture of surface layers in atherosclerotic plaque can cause heart attack and stroke; however, changes in luminal surface composition are incompletely defined. Laser-induced fluorescence spectroscopy (LIFS), with limited tissue penetration, was used to investigate the surface of unstable carotid plaque and correlated with microscopy, birefringence and gene expression. Arterial matrix collagens I, III and elastin were assessed in unstable plaques (n = 25) and reference left internal mammary arteries (LIMA, n = 10). LIFS in addition to selective histological staining with picrosirius red, Movat pentachrome and immunostaining revealed decreased elastin and increased collagen I and III (P < 0.05) in carotid plaque when compared with LIMA. Within plaque, collagen I was elevated in the internal carotid region versus the common carotid region. Polarized light microscopy detected layers of aligned collagen and associated mechanical rigidity of the fibrous cap. Microarray analysis of three carotid and three LIMA specimens confirmed up-regulation of collagen I, III and IV, lysyl oxidase and MMP-12. In conclusion, LIFS analysis coupled with microscopy revealed marked regional differences in collagen I, III and elastin in surface layers of carotid plaque; indicative of plaque instability. Birefringence measurements demonstrated mechanical rigidity and weakening of the fibrous cap with complementary changes in ECM gene expression.


Asunto(s)
Arterias Carótidas/metabolismo , Estenosis Carotídea/metabolismo , Matriz Extracelular/química , Placa Aterosclerótica/metabolismo , Anciano , Anciano de 80 o más Años , Birrefringencia , Arterias Carótidas/patología , Arterias Carótidas/cirugía , Estenosis Carotídea/patología , Estenosis Carotídea/cirugía , Elastina/genética , Elastina/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Colágenos Fibrilares/genética , Colágenos Fibrilares/metabolismo , Colorantes Fluorescentes/análisis , Humanos , Inmunohistoquímica , Rayos Láser , Masculino , Metaloproteinasa 12 de la Matriz/genética , Metaloproteinasa 12 de la Matriz/metabolismo , Persona de Mediana Edad , Placa Aterosclerótica/patología , Placa Aterosclerótica/cirugía , Análisis por Matrices de Proteínas , Proteína-Lisina 6-Oxidasa/genética , Proteína-Lisina 6-Oxidasa/metabolismo , Espectrometría de Fluorescencia
11.
PLoS One ; 5(12): e15132, 2010 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-21151997

RESUMEN

Membrane-associated RING-CH (MARCH) proteins represent a family of transmembrane ubiquitin ligases modulating intracellular trafficking and turnover of transmembrane protein targets. While homologous proteins encoded by gamma-2 herpesviruses and leporipoxviruses have been studied extensively, limited information is available regarding the physiological targets of cellular MARCH proteins. To identify host cell proteins targeted by the human MARCH-VIII ubiquitin ligase we used stable isotope labeling of amino-acids in cell culture (SILAC) to monitor MARCH-dependent changes in the membrane proteomes of human fibroblasts. Unexpectedly, we observed that MARCH-VIII reduced the surface expression of Bap31, a chaperone that predominantly resides in the endoplasmic reticulum (ER). We demonstrate that Bap31 associates with the transmembrane domains of several MARCH proteins and controls intracellular transport of MARCH proteins. In addition, we observed that MARCH-VIII reduced the surface expression of the hyaluronic acid-receptor CD44 and both MARCH-VIII and MARCH-IV sequestered the tetraspanin CD81 in endo-lysosomal vesicles. Moreover, gene knockdown of MARCH-IV increased surface levels of endogenous CD81 suggesting a constitutive involvement of this family of ubiquitin ligases in the turnover of tetraspanins. Our data thus suggest a role of MARCH-VIII and MARCH-IV in the regulated turnover of CD81 and CD44, two ubiquitously expressed, multifunctional proteins.


Asunto(s)
Antígenos CD/metabolismo , Receptores de Hialuranos/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Membrana Celular/metabolismo , Endosomas/metabolismo , Fibroblastos/metabolismo , Células HeLa , Humanos , Ácido Hialurónico/química , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Proteoma , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Tetraspanina 28 , Tetraspanina 29
12.
Curr Opin Microbiol ; 13(4): 517-23, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20699190

RESUMEN

The post-translational attachment of ubiquitin or ubiquitin-like modifiers (ULMs) to proteins regulates many cellular processes including the generation of innate and adaptive immune responses to pathogens. Vice versa, pathogens counteract immune defense by inhibiting or redirecting the ubiquitination machinery of the host. A common immune evasion strategy is for viruses to target host immunoproteins for proteasomal or lysosomal degradation by employing viral or host ubiquitin ligases. By degrading key host adaptor and signaling molecules, viruses thus disable multiple immune response pathways including the production of and response to interferons as well as other innate host defense mechanisms. Recent work further revealed that viruses inhibit the ligation of ubiquitin or ULMs or remove ubiquitin from host cell proteins. Thus, viruses succeed in either stabilizing negative regulators of innate immune signaling or thwart host cell proteins that are activated by ubiquitin or ULM-modification.


Asunto(s)
Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Virosis/inmunología , Virosis/virología , Virus/inmunología , Humanos , Evasión Inmune , Inmunidad Innata , Interferones/inmunología , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Virosis/enzimología
13.
PLoS Pathog ; 6(5): e1000913, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20485522

RESUMEN

The interferon-induced BST-2 protein has the unique ability to restrict the egress of HIV-1, Kaposi's sarcoma-associated herpesvirus (KSHV), Ebola virus, and other enveloped viruses. The observation that virions remain attached to the surface of BST-2-expressing cells led to the renaming of BST-2 as "tetherin". However, viral proteins such as HIV-1 Vpu, simian immunodeficiency virus Nef, and KSHV K5 counteract BST-2, thereby allowing mature virions to readily escape from infected cells. Since the anti-viral function of BST-2 was discovered, there has been an explosion of research into several aspects of this intriguing interplay between host and virus. This review focuses on recent work addressing the molecular mechanisms involved in BST-2 restriction of viral egress and the species-specific countermeasures employed by various viruses.


Asunto(s)
Antígenos CD/metabolismo , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/crecimiento & desarrollo , Glicoproteínas de Membrana/metabolismo , Antígenos CD/genética , Proteínas Ligadas a GPI , Humanos , Glicoproteínas de Membrana/genética , Virión/metabolismo , Virosis/metabolismo , Virosis/virología
14.
J Virol ; 83(19): 9672-81, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19605472

RESUMEN

K3/MIR1 and K5/MIR2 of Kaposi's sarcoma-associated herpesvirus (KSHV) are viral members of the membrane-associated RING-CH (MARCH) ubiquitin ligase family and contribute to viral immune evasion by directing the conjugation of ubiquitin to immunostimulatory transmembrane proteins. In a quantitative proteomic screen for novel host cell proteins downregulated by viral immunomodulators, we previously observed that K5, as well as the human immunodeficiency virus type 1 (HIV-1) immunomodulator VPU, reduced steady-state levels of bone marrow stromal cell antigen 2 (BST2; also called CD317 or tetherin), suggesting that BST2 might be a novel substrate of K5 and VPU. Recent work revealed that in the absence of VPU, HIV-1 virions are tethered to the plasma membrane in BST2-expressing HeLa cells. By targeting BST2, K5 might thus similarly overcome an innate antiviral host defense mechanism. Here we establish that despite its type II transmembrane topology and carboxy-terminal glycosylphosphatidylinositol (GPI) anchor, BST2 represents a bona fide target of K5 that is downregulated during primary infection by and reactivation of KSHV. Upon exit of the protein from the endoplasmic reticulum, lysines in the short amino-terminal domain of BST2 are ubiquitinated by K5, resulting in rapid degradation of BST2. Ubiquitination of BST2 is required for degradation, since BST2 lacking cytosolic lysines was K5 resistant and ubiquitin depletion by proteasome inhibitors restored BST2 surface expression. Thus, BST2 represents the first type II transmembrane protein targeted by K5 and the first example of a protein that is both ubiquitinated and GPI linked. We further demonstrate that KSHV release is decreased in the absence of K5 in a BST2-dependent manner, suggesting that K5 contributes to the evasion of intracellular antiviral defense programs.


Asunto(s)
Antígenos CD/biosíntesis , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/metabolismo , Proteínas Inmediatas-Precoces/biosíntesis , Glicoproteínas de Membrana/biosíntesis , Proteínas Virales/biosíntesis , Biotinilación , Células Cultivadas , Células Endoteliales/virología , Proteínas Ligadas a GPI , Células HeLa , Humanos , Microcirculación , Modelos Biológicos , Reacción en Cadena de la Polimerasa , Complejo de la Endopetidasa Proteasomal/metabolismo
15.
J Virol ; 83(16): 7931-47, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19515779

RESUMEN

The primary roles attributed to the human immunodeficiency virus type 1 (HIV-1) Vpu protein are the degradation of the viral receptor CD4 and the enhancement of virion release. With regard to CD4 downregulation, Vpu has been shown to act as an adapter linking CD4 with the ubiquitin-proteasome machinery via interaction with the F-box protein betaTrCP. To identify additional cellular betaTrCP-dependent Vpu targets, we performed quantitative proteomics analyses using the plasma membrane fraction of HeLa cells expressing either wild-type Vpu or a Vpu mutant (S52N/S56N) that does not bind betaTrCP. One cellular protein, BST-2 (CD317), was consistently underrepresented in the membrane proteome of cells expressing wild-type Vpu compared to the proteome of cells expressing the Vpu mutant. To verify the biological relevance of this phenotype for HIV pathogenesis, we showed that in T cells infected with HIV-1, BST-2 downregulation occurred in a Vpu-dependent manner. Recently, BST-2 has been identified as the interferon-inducible cellular factor Tetherin, which restricts HIV virion release in the absence of Vpu. We address here the unresolved mechanism of Vpu-mediated BST-2 downregulation. Our data show that the presence of wild-type Vpu reduced cell surface and total steady-state BST-2 levels, whereas that of the mutant Vpu had no effect. In addition, treatment of cells with the lysosome acidification inhibitor concanamycin A, but not treatment with the proteasome inhibitor MG132, reduced BST-2 downregulation by wild-type Vpu, thereby suggesting that the presence of Vpu leads to the degradation of BST-2 via an endosome-lysosome degradation pathway. The importance of betaTrCP in this process was confirmed by demonstrating that in the absence of betaTrCP, BST-2 levels were restored despite the presence of Vpu. Taken together, these data support the hypothesis that, in similarity to its role in CD4 degradation, Vpu acts as an adapter molecule linking BST-2 to the cellular ubiquitination machinery via betaTrCP. However, in contrast to the proteasome-dependent degradation of CD4, which occurs in the endoplasmic reticulum, Vpu appears to interact with BST-2 in the trans-Golgi network or in early endosomes, leading to lysosomal degradation of BST-2. Via this action, Vpu could counter the tethering function of BST-2, resulting in enhanced HIV-1 virion release. Interestingly, although HIV-2 does not express Vpu, an isolate known to exhibit enhanced viral egress can downregulate surface BST-2 by an as-yet-unknown mechanism that does not appear to involve degradation. Understanding the molecular mechanisms of both Vpu-dependent and -independent mediated antagonism of BST-2 will be critical for therapeutic strategies that exploit this novel viral function.


Asunto(s)
Antígenos CD/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas Reguladoras y Accesorias Virales/fisiología , Proteínas con Repetición de beta-Transducina/metabolismo , Antígenos CD/genética , Antígenos CD4/genética , Antígenos CD4/metabolismo , Línea Celular , Proteínas Ligadas a GPI , Infecciones por VIH/virología , VIH-1/genética , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Glicoproteínas de Membrana/genética , Unión Proteica , Transporte de Proteínas , Proteínas Reguladoras y Accesorias Virales/genética , Virión/genética , Virión/fisiología , Esparcimiento de Virus , Proteínas con Repetición de beta-Transducina/genética
16.
Adv Exp Med Biol ; 666: 132-56, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20054981

RESUMEN

Serine proteinase inhibitors, also called serpins, are an ancient grouping of proteins found in primitive organisms from bacteria, protozoa and horseshoe crabs and thus likely present at the time of the dinosaurs, up to all mammals living today. The innate or inflammatory immune system is also an ancient metazoan regulatory system, providing the first line of defense against infection or injury. The innate inflammatory defense response evolved long before acquired, antibody dependent immunity. Viruses have developed highly effective stratagems that undermine and block a wide variety of host inflammatory and immune responses. Some of the most potent of these immune modifying strategies utilize serpins that have also been developed over millions of years, including the hijacking by some viruses for defense against host immune attacks. Serpins represent up to 2-10 percent of circulating plasma proteins, regulating actions as wide ranging as thrombosis, inflammation, blood pressure control and even hormone transport. Targeting serpin-regulated immune or inflammatory pathways makes evolutionary sense for viral defense and many of these virus-derived inhibitory proteins have proven to be highly effective, working at very low concentrations--even down to the femptomolar to picomolar range. We are studying these viral anti-inflammatory proteins as a new class of immunomodulatory therapeutic agents derived from their native viral source. One such viral serpin, Serp-1 is now in clinical trial (conducted by VIRON Therapeutics, Inc.) for acute unstable coronary syndromes (unstable angina and small heart attacks), representing a 'first in class' therapeutic study. Several other viral serpins are also currently under investigation as anti-inflammatory or anti-immune therapeutics. This chapter describes these original studies and the ongoing analysis of viral serpins as a new class of virus-derived immunotherapeutic.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedades del Sistema Inmune/terapia , Inhibidores de Serina Proteinasa/uso terapéutico , Serpinas/uso terapéutico , Proteínas Virales/uso terapéutico , Virosis/terapia , Animales , Humanos
17.
J Leukoc Biol ; 85(3): 418-26, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19052145

RESUMEN

Serp-1 is a secreted myxoma viral serine protease inhibitor (serpin) with proven, highly effective, anti-inflammatory defensive activity during host cell infection, as well as potent immunomodulatory activity in a wide range of animal disease models. Serp-1 binds urokinase-type plasminogen activator (uPA) and the tissue-type PA, plasmin, and factor Xa, requiring uPA receptor (uPAR) for anti-inflammatory activity. To define Serp-1-mediated effects on inflammatory cell activation, we examined the association of Serp-1 with monocytes and T cells, effects on cellular migration, and the role of uPAR-linked integrins and actin-binding proteins in Serp-1 cellular responses. Our results show that Serp-1 associates directly with activated monocytes and T lymphocytes, in part through interaction with uPAR (P<0.001). Serp-1, but not mammalian serpin PA inhibitor-1 (PAI-1), attenuated cellular adhesion to the extracellular matrix. Serp-1 and PAI-1 reduced human monocyte and T cell adhesion (P<0.001) and migration across endothelial monolayers in vitro (P<0.001) and into mouse ascites in vivo (P<0.001). Serp-1 and an inactive Serp-1 mutant Serp-1(SAA) bound equally to human monocytes and T cells, but a highly proinflammatory mutant, Serp-1(Ala(6)), bound less well to monocytes. Serp-1 treatment of monocytes increased expression of filamin B actin-binding protein and reduced CD18 (beta-integrin) expression (P<0.001) in a uPAR-dependent response. Filamin colocalized and co-immunoprecipitated with uPAR, and short interference RNA knock-down of filamin blocked Serp-1 inhibition of monocyte adhesion. We report here that the highly potent, anti-inflammatory activity of Serp-1 is mediated through modification of uPAR-linked beta-integrin and filamin in monocytes, identifying this interaction as a central regulatory axis for inflammation.


Asunto(s)
Adhesión Celular , Proteínas Contráctiles/metabolismo , Proteínas de Microfilamentos/metabolismo , Monocitos/citología , Myxoma virus/patogenicidad , Serpinas/fisiología , Proteínas Virales/fisiología , Filaminas , Humanos , Inflamación , Cadenas beta de Integrinas/metabolismo , Unión Proteica/inmunología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
18.
Expert Rev Proteomics ; 4(6): 815-29, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18067418

RESUMEN

Viruses constantly adapt to and modulate the host environment during replication and propagation. Both DNA and RNA viruses encode multifunctional proteins that interact with and modify host cell proteins. While viral genomes were the first complete sequences known, the corresponding proteomes are only now elucidated, with some surprising results. Even more daunting is the task to globally monitor the impact of viral infection on the proteome of the host cell and many technical hurdles must still be overcome in order to facilitate robust and reproducible measurements. Further complicating the picture is the dynamic nature of proteins, including post-translational modifications, enzymatic cleavage and activation or destruction by proteolytic events. Nevertheless, several promising studies have been published using high-throughput methods directly measuring protein abundance. Particularly, quantitative or semiquantitative mass spectrometry-based analysis of viral and cellular proteomes are now being used to characterize viruses and their host interaction. In addition, the full set of interactions between viral and host proteins, the interactome, is beginning to emerge, with often unexpected interactions that need to be carefully validated. In this review, we will discuss two major areas of viral proteomics: first, virion proteomics (such as the protein characterization of viral particles) and second, proteoviromics, including the viral protein interactomics and the quantitative analysis of host cell proteome during viral infection.


Asunto(s)
Genes Virales , Proteoma/genética , Proteómica/métodos , Virosis/metabolismo , Virosis/virología , Virus/genética , Virus/metabolismo , ADN/química , Electroforesis en Gel Bidimensional , Espectrometría de Masas/métodos , Proteoma/química , ARN/química , Técnicas del Sistema de Dos Híbridos , Proteínas Virales/química , Virión , Fenómenos Fisiológicos de los Virus
19.
Cardiovasc Pathol ; 16(4): 191-202, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17637427

RESUMEN

BACKGROUND: Angiogenesis is a critical factor in the development of malignant tumors, in arthritic joints, and in cardiovascular disease. In cardiovascular disease, angiogenesis is recognised both as a potential therapy and as a complicating factor in atherosclerotic plaque rupture and thrombotic obstruction. Serine proteases regulate thrombosis, inflammation, and cell invasion, events that trigger various stages of angiogenesis and are in turn regulated by inhibitors, termed serpins. Serp-1 is a secreted anti-inflammatory viral serpin that profoundly inhibits early mononuclear cell invasion, and the development of atherosclerosis, transplant vasculopathy, and arthritis in a range of animal models. METHODS: The capacity of Serp-1 to alter angiogenesis was evaluated in the chicken chorioallantoic membrane (CAM) model using morphometric analysis of vascular changes and RT-PCR to explore alterations in gene expression. RESULTS: Serp-1 inhibited endogenous angiogenesis in a dose-dependent manner, with associated altered expression of laminin and vascular endothelial growth factor (VEGF). Serp-1 was ineffective in CAMs no longer in the rapid growth phase. Similar inhibition of angiogenesis was detected after inhibition of VEGF, but not after treatment with the inactivated reactive center loop mutant of Serp-1. CONCLUSIONS: The angiogenic process can be controlled using Serp-1, an anti-inflammatory agent that is effective at low concentrations with rapid reversibility, targets endothelial cells, and reduces the availability of VEGF. These properties may be especially important in cardiovascular disease, reducing plaque destabilization. It is likely that the anti-angiogenic activity of Serp-1 contributes to the observed anti-inflammatory and anti-atherogenic actions with potential importance in this therapeutic setting.


Asunto(s)
Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Serpinas/farmacología , Proteínas Virales/farmacología , Animales , Embrión de Pollo , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Laminina/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
20.
Atherosclerosis ; 194(1): 116-24, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17126342

RESUMEN

OBJECTIVE: Apolipoprotein E (ApoE) is a lipid transport protein with expanded functions in cellular responses to tissue injury, immune regulation and cell growth. ApoE directs vascular changes that contribute to arterial protection as evidenced by the fact that isoforms of ApoE and ApoE deficiency correlate closely with accelerated plaque growth. The N-terminus of the ApoE protein has well-characterized functions, displaying lipid-binding and anti-atherogenic activity, whereas the function of the C-terminus is only partially defined. We have assessed the effects of a 14 amino acid C-terminal ApoE peptide, termed Ep1.B (239-252), on intimal neoplasia in animal models. This peptide is a fragment of a naturally processed peptide (236-252) of murine ApoE. METHODS AND RESULTS: Ep1.B injection reduced neointimal hyperplasia after vascular surgery in rats and mice. When given during early plaque progression in ApoE-deficient mice, Ep1.B injections also prevented plaque growth. Treatment with Ep1.B did not, however, reduce established plaque growth in older mice. Peptides with alanine substitution of amino acid 249, Ep1.N, and with complete sequence reversal, Ep1.R, did not consistently inhibit plaque growth. CONCLUSION: A naturally processed C-terminal ApoE peptide, Ep1.B, has anti-atherogenic activity indicating a role for this naturally metabolized peptide in vascular wound healing and lipid homeostasis.


Asunto(s)
Apolipoproteínas E/farmacología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Fragmentos de Péptidos/farmacología , Angioplastia/efectos adversos , Animales , Apolipoproteínas E/síntesis química , Apolipoproteínas E/química , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/inmunología , Traumatismos de las Arterias Carótidas/inmunología , Traumatismos de las Arterias Carótidas/patología , Modelos Animales de Enfermedad , Femenino , Arteria Femoral/inmunología , Arteria Femoral/lesiones , Arteria Femoral/patología , Antígenos de Histocompatibilidad Clase II/metabolismo , Hiperplasia , Arteria Ilíaca/lesiones , Arteria Ilíaca/patología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/metabolismo , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Linfocitos T/patología , Túnica Íntima/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...