Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
PLoS Genet ; 19(7): e1010834, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37418503

RESUMEN

Sulfur is an indispensable element for bacterial proliferation. Prior studies demonstrated that the human pathogen Staphylococcus aureus utilizes glutathione (GSH) as a source of nutrient sulfur; however, mechanisms of GSH acquisition are not defined. Here, we identify a five-gene locus comprising a putative ABC-transporter and predicted γ-glutamyl transpeptidase (ggt) that promotes S. aureus proliferation in medium supplemented with either reduced or oxidized GSH (GSSG) as the sole source of nutrient sulfur. Based on these phenotypes, we name this transporter operon the glutathione import system (gisABCD). Ggt is encoded within the gisBCD operon, and we show that the enzyme is capable of liberating glutamate using either GSH or GSSG as substrates, demonstrating it is a bona fide γ-glutamyl transpeptidase. We also determine that Ggt is expressed in the cytoplasm, representing only the second example of cytoplasmic Ggt localization, the other being Neisseria meningitidis. Bioinformatic analyses revealed that Staphylococcus species closely related to S. aureus encode GisABCD-Ggt homologs. However, homologous systems were not detected in Staphylococcus epidermidis. Consequently, we establish that GisABCD-Ggt provides a competitive advantage for S. aureus over S. epidermidis in a GSH- and GSSG-dependent manner. Overall, this study describes the discovery of a nutrient sulfur acquisition system in S. aureus that targets GSSG in addition to GSH and promotes competition against other staphylococci commonly associated with the human microbiota.


Asunto(s)
Staphylococcus aureus , gamma-Glutamiltransferasa , Humanos , Staphylococcus aureus/genética , gamma-Glutamiltransferasa/genética , Disulfuro de Glutatión , Glutatión/genética , Azufre
2.
Infect Immun ; 88(3)2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31843961

RESUMEN

Staphylococcus aureus is a significant human pathogen due to its capacity to cause a multitude of diseases. As such, S. aureus efficiently pillages vital nutrients from the host; however, the molecular mechanisms that support sulfur acquisition during infection have not been established. One of the most abundant extracellular sulfur-containing metabolites within the host is cysteine, which acts as the major redox buffer in the blood by transitioning between reduced and oxidized (cystine) forms. We therefore hypothesized that S. aureus acquires host-derived cysteine and cystine as sources of nutrient sulfur during systemic infection. To test this hypothesis, we used the toxic cystine analogue selenocystine to initially characterize S. aureus homologues of the Bacillus subtilis cystine transporters TcyABC and TcyP. We found that genetic inactivation of both TcyA and TcyP induced selenocystine resistance. The double mutant also failed to proliferate in medium supplemented with cystine, cysteine, or N-acetyl cysteine as the sole sulfur source. However, only TcyABC was necessary for proliferation in defined medium containing homocystine as the sulfur source. Using a murine model of systemic infection, we observed tcyP-dependent competitive defects in the liver and heart, indicating that this sulfur acquisition strategy supports proliferation of S. aureus in these organs. Phylogenetic analyses identified TcyP homologues in many pathogenic species, implying that this sulfur procurement strategy is conserved. In total, this study is the first to experimentally validate sulfur acquisition systems in S. aureus and establish their importance during pathogenesis.


Asunto(s)
Cistina/metabolismo , Proteínas de Transporte de Membrana/fisiología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/fisiología , Azufre/metabolismo , Animales , Ratones
3.
J Bacteriol ; 200(11)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29581406

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) is a threat to global health. Consequently, much effort has focused on the development of new antimicrobials that target novel aspects of S. aureus physiology. Fatty acids are required to maintain cell viability, and bacteria synthesize fatty acids using the type II fatty acid synthesis (FASII) pathway. FASII is significantly different from human fatty acid synthesis, underscoring the therapeutic potential of inhibiting this pathway. However, many Gram-positive pathogens incorporate exogenous fatty acids, bypassing FASII inhibition and leaving the clinical potential of FASII inhibitors uncertain. Importantly, the source(s) of fatty acids available to pathogens within the host environment remains unclear. Fatty acids are transported throughout the body by lipoprotein particles in the form of triglycerides and esterified cholesterol. Thus, lipoproteins, such as low-density lipoprotein (LDL), represent a potentially rich source of exogenous fatty acids for S. aureus during infection. We sought to test the ability of LDLs to serve as a fatty acid source for S. aureus and show that cells cultured in the presence of human LDLs demonstrate increased tolerance to the FASII inhibitor triclosan. Using mass spectrometry, we observed that host-derived fatty acids present in the LDLs are incorporated into the staphylococcal membrane and that tolerance to triclosan is facilitated by the fatty acid kinase A, FakA, and Geh, a triacylglycerol lipase. Finally, we demonstrate that human LDLs support the growth of S. aureus fatty acid auxotrophs. Together, these results suggest that human lipoprotein particles are a viable source of exogenous fatty acids for S. aureus during infection.IMPORTANCE Inhibition of bacterial fatty acid synthesis is a promising approach to combating infections caused by S. aureus and other human pathogens. However, S. aureus incorporates exogenous fatty acids into its phospholipid bilayer. Therefore, the clinical utility of targeting bacterial fatty acid synthesis is debated. Moreover, the fatty acid reservoir(s) exploited by S. aureus is not well understood. Human low-density lipoprotein particles represent a particularly abundant in vivo source of fatty acids and are present in tissues that S. aureus colonizes. Herein, we establish that S. aureus is capable of utilizing the fatty acids present in low-density lipoproteins to bypass both chemical and genetic inhibition of fatty acid synthesis. These findings imply that S. aureus targets LDLs as a source of fatty acids during pathogenesis.


Asunto(s)
Ácidos Grasos/biosíntesis , Lipoproteínas/metabolismo , Staphylococcus aureus Resistente a Meticilina/metabolismo , Infecciones Estafilocócicas/microbiología , Triclosán/metabolismo , Farmacorresistencia Bacteriana , Humanos , Lipoproteínas LDL/metabolismo , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Mutación , Fosfolípidos/metabolismo
4.
J Virol ; 88(19): 11199-214, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25031353

RESUMEN

Neurotropic alphaviruses, including western, eastern, and Venezuelan equine encephalitis viruses, cause serious and potentially fatal central nervous system infections in humans for which no currently approved therapies exist. We previously identified a series of thieno[3,2-b]pyrrole derivatives as novel inhibitors of neurotropic alphavirus replication, using a cell-based phenotypic assay (W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275), and subsequently developed second- and third-generation indole-2-carboxamide derivatives with improved potency, solubility, and metabolic stability (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). In this report, we describe the antiviral activity of the most promising third-generation lead compound, CCG205432, and closely related analogs CCG206381 and CCG209023. These compounds have half-maximal inhibitory concentrations of ∼1 µM and selectivity indices of >100 in cell-based assays using western equine encephalitis virus replicons. Furthermore, CCG205432 retains similar potency against fully infectious virus in cultured human neuronal cells. These compounds show broad inhibitory activity against a range of RNA viruses in culture, including members of the Togaviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Although their exact molecular target remains unknown, mechanism-of-action studies reveal that these novel indole-based compounds target a host factor that modulates cap-dependent translation. Finally, we demonstrate that both CCG205432 and CCG209023 dampen clinical disease severity and enhance survival of mice given a lethal western equine encephalitis virus challenge. These studies demonstrate that indole-2-carboxamide compounds are viable candidates for continued preclinical development as inhibitors of neurotropic alphaviruses and, potentially, of other RNA viruses. IMPORTANCE There are currently no approved drugs to treat infections with alphaviruses. We previously identified a novel series of compounds with activity against these potentially devastating pathogens (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). We have now produced third-generation compounds with enhanced potency, and this manuscript provides detailed information on the antiviral activity of these advanced-generation compounds, including activity in an animal model. The results of this study represent a notable achievement in the continued development of this novel class of antiviral inhibitors.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Equina del Oeste/efectos de los fármacos , Encefalomielitis Equina/tratamiento farmacológico , Indoles/farmacología , Piridinas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/síntesis química , Bunyaviridae/efectos de los fármacos , Bunyaviridae/crecimiento & desarrollo , Línea Celular , Virus de la Encefalitis Equina del Oeste/crecimiento & desarrollo , Virus de la Encefalitis Equina del Oeste/patogenicidad , Encefalomielitis Equina/mortalidad , Encefalomielitis Equina/virología , Femenino , Indoles/síntesis química , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/virología , Paramyxoviridae/efectos de los fármacos , Paramyxoviridae/crecimiento & desarrollo , Picornaviridae/efectos de los fármacos , Picornaviridae/crecimiento & desarrollo , Biosíntesis de Proteínas/efectos de los fármacos , Piridinas/síntesis química , Replicón/efectos de los fármacos , Relación Estructura-Actividad , Análisis de Supervivencia
5.
J Neurovirol ; 21(1): 43-55, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25361697

RESUMEN

While alphaviruses spread naturally via mosquito vectors, some can also be transmitted as aerosols making them potential bioterrorism agents. One such pathogen, western equine encephalitis virus (WEEV), causes fatal human encephalitis via multiple routes of infection and thus presumably via multiple mechanisms. Although WEEV also produces acute encephalitis in non-human primates, a small animal model that recapitulates features of human disease would be useful for both pathogenesis studies and to evaluate candidate antiviral therapies. We have optimized conditions to infect mice with a low passage isolate of WEEV, thereby allowing detailed investigation of virus tropism, replication, neuroinvasion, and neurovirulence. We find that host factors strongly influence disease outcome, and in particular, that age, gender, and genetic background all have significant effects on disease susceptibility independent of virus tropism or replication within the central nervous system. Our data show that experimental variables can be adjusted in mice to recapitulate disease features known to occur in both non-human primates and humans, thus aiding further study of WEEV pathogenesis and providing a realistic therapeutic window for antiviral drug delivery.


Asunto(s)
Infecciones por Alphavirus/patología , Virus de la Encefalitis Equina del Oeste/patogenicidad , ARN Viral/sangre , Convulsiones/patología , Administración Intranasal , Infecciones por Alphavirus/virología , Animales , Conducta Animal , Cognición , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina del Oeste/fisiología , Especificidad del Huésped , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Convulsiones/virología , Especificidad de la Especie , Carga Viral , Replicación Viral
6.
J Biol Chem ; 285(53): 41432-42, 2010 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-21041303

RESUMEN

Thrombin is a potent modulator of endothelial function and, through stimulation of NF-κB, induces endothelial expression of intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). These cell surface adhesion molecules recruit inflammatory cells to the vessel wall and thereby participate in the development of atherosclerosis, which is increasingly recognized as an inflammatory condition. The principal receptor for thrombin on endothelial cells is protease-activated receptor-1 (PAR-1), a member of the G protein-coupled receptor superfamily. Although it is known that PAR-1 signaling to NF-κB depends on initial PKC activation, the subsequent steps leading to stimulation of the canonical NF-κB machinery have remained unclear. Here, we demonstrate that a complex of proteins containing CARMA3, Bcl10, and MALT1 links PAR-1 activation to stimulation of the IκB kinase complex. IκB kinase in turn phosphorylates IκB, leading to its degradation and the release of active NF-κB. Further, we find that although this CARMA3·Bcl10·MALT1 signalosome shares features with a CARMA1-containing signalosome found in lymphocytes, there are significant differences in how the signalosomes communicate with their cognate receptors. Specifically, whereas the CARMA1-containing lymphocyte complex relies on 3-phosphoinositide-dependent protein kinase 1 for assembly and activation, the CARMA3-containing endothelial signalosome functions completely independent of 3-phosphoinositide-dependent protein kinase 1 and instead relies on ß-arrestin 2 for assembly. Finally, we show that thrombin-dependent adhesion of monocytes to endothelial cells requires an intact endothelial CARMA3·Bcl10·MALT1 signalosome, underscoring the importance of the signalosome in mediating one of the most significant pro-atherogenic effects of thrombin.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasas/metabolismo , Células Endoteliales/citología , Regulación de la Expresión Génica , Monocitos/citología , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Proteinasa-Activados/metabolismo , Trombina/química , Trombina/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Proteína 10 de la LLC-Linfoma de Células B , Humanos , Inflamación , Ratones , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Transducción de Señal
7.
J Biol Chem ; 285(34): 25880-4, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20605784

RESUMEN

The CARMA1, Bcl10, and MALT1 proteins together constitute a signaling complex (CBM signalosome) that mediates antigen-dependent activation of NF-kappaB in lymphocytes, thereby representing a cornerstone of the adaptive immune response. Although CARMA1 is restricted to cells of the immune system, the analogous CARMA3 protein has a much wider expression pattern. Emerging evidence suggests that CARMA3 can substitute for CARMA1 in non-immune cells to assemble a CARMA3-Bcl10-MALT1 signalosome and mediate G protein-coupled receptor activation of NF-kappaB. Here we show that one G protein-coupled receptor, the type 1 receptor for angiotensin II, utilizes this mechanism for activation of NF-kappaB in endothelial and vascular smooth muscle cells, thereby inducing pro-inflammatory signals within the vasculature, a key factor in atherogenesis. Further, we demonstrate that Bcl10-deficient mice are protected from developing angiotensin-dependent atherosclerosis and aortic aneurysms. By uncovering a novel vascular role for the CBM signalosome, these findings illustrate that CBM-dependent signaling has functions outside the realm of adaptive immunity and impacts pathobiology more broadly than previously known.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Angiotensina II/fisiología , Aterosclerosis/etiología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasas/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal/fisiología , Animales , Aterosclerosis/patología , Proteína 10 de la LLC-Linfoma de Células B , Vasos Sanguíneos/patología , Endotelio Vascular/patología , Inflamación/etiología , Ratones , Ratones Noqueados , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Músculo Liso Vascular/patología , FN-kappa B/metabolismo , Receptor de Angiotensina Tipo 1
8.
J Clin Invest ; 130(2): 1036-1051, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31961340

RESUMEN

Antigen receptor-dependent (AgR-dependent) stimulation of the NF-κB transcription factor in lymphocytes is a required event during adaptive immune response, but dysregulated activation of this signaling pathway can lead to lymphoma. AgR stimulation promotes assembly of the CARMA1-BCL10-MALT1 complex, wherein MALT1 acts as (a) a scaffold to recruit components of the canonical NF-κB machinery and (b) a protease to cleave and inactivate specific substrates, including negative regulators of NF-κB. In multiple lymphoma subtypes, malignant B cells hijack AgR signaling pathways to promote their own growth and survival, and inhibiting MALT1 reduces the viability and growth of these tumors. As such, MALT1 has emerged as a potential pharmaceutical target. Here, we identified G protein-coupled receptor kinase 2 (GRK2) as a new MALT1-interacting protein. We demonstrated that GRK2 binds the death domain of MALT1 and inhibits MALT1 scaffolding and proteolytic activities. We found that lower GRK2 levels in activated B cell-type diffuse large B cell lymphoma (ABC-DLBCL) are associated with reduced survival, and that GRK2 knockdown enhances ABC-DLBCL tumor growth in vitro and in vivo. Together, our findings suggest that GRK2 can function as a tumor suppressor by inhibiting MALT1 and provide a roadmap for developing new strategies to inhibit MALT1-dependent lymphomagenesis.


Asunto(s)
Carcinogénesis/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Proteínas Oncogénicas/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Femenino , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Humanos , Células Jurkat , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Endogámicos NOD , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , Proteínas Oncogénicas/genética
9.
J Vis Exp ; (147)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31157784

RESUMEN

Staphylococcus aureus and other Gram-positive pathogens incorporate fatty acids from the environment into membrane phospholipids. During infection, the majority of exogenous fatty acids are present within host lipoprotein particles. Uncertainty remains as to the reservoirs of host fatty acids and the mechanisms by which bacteria extract fatty acids from the lipoprotein particles. In this work, we describe protocols for enrichment of low-density lipoprotein (LDL) particles from chicken egg yolk and determining whether LDLs serve as fatty acid reservoirs for S. aureus. This method exploits unbiased lipidomic analysis and chicken LDLs, an effective and economical model for the exploration of interactions between LDLs and bacteria. The analysis of S. aureus integration of exogenous fatty acids from LDLs is performed using high-resolution/accurate mass spectrometry and tandem mass spectrometry, enabling the characterization of the fatty acid composition of the bacterial membrane and unbiased identification of novel combinations of fatty acids that arise in bacterial membrane lipids upon exposure to LDLs. These advanced mass spectrometry techniques offer an unparalleled perspective of fatty acid incorporation by revealing the specific exogenous fatty acids incorporated into the phospholipids. The methods outlined here are adaptable to the study of other bacterial pathogens and alternative sources of complex fatty acids.


Asunto(s)
Pollos/metabolismo , Yema de Huevo/metabolismo , Ácidos Grasos/metabolismo , Lipoproteínas LDL/aislamiento & purificación , Fosfolípidos/metabolismo , Staphylococcus aureus/metabolismo , Animales , Membranas/metabolismo , Fosfolípidos/química
10.
Cancer Res ; 78(5): 1225-1240, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29259013

RESUMEN

The angiotensin II receptor AGTR1, which mediates vasoconstrictive and inflammatory signaling in vascular disease, is overexpressed aberrantly in some breast cancers. In this study, we established the significance of an AGTR1-responsive NFκB signaling pathway in this breast cancer subset. We documented that AGTR1 overexpression occurred in the luminal A and B subtypes of breast cancer, was mutually exclusive of HER2 expression, and correlated with aggressive features that include increased lymph node metastasis, reduced responsiveness to neoadjuvant therapy, and reduced overall survival. Mechanistically, AGTR1 overexpression directed both ligand-independent and ligand-dependent activation of NFκB, mediated by a signaling pathway that requires the triad of CARMA3, Bcl10, and MALT1 (CBM signalosome). Activation of this pathway drove cancer cell-intrinsic responses that include proliferation, migration, and invasion. In addition, CBM-dependent activation of NFκB elicited cancer cell-extrinsic effects, impacting endothelial cells of the tumor microenvironment to promote tumor angiogenesis. CBM/NFκB signaling in AGTR1+ breast cancer therefore conspires to promote aggressive behavior through pleiotropic effects. Overall, our results point to the prognostic and therapeutic value of identifying AGTR1 overexpression in a subset of HER2-negative breast cancers, and they provide a mechanistic rationale to explore the repurposing of drugs that target angiotensin II-dependent NFκB signaling pathways to improve the treatment of this breast cancer subset.Significance: These findings offer a mechanistic rationale to explore the repurposing of drugs that target angiotensin action to improve the treatment of AGTR1-expressing breast cancers. Cancer Res; 78(5); 1225-40. ©2017 AACR.


Asunto(s)
Proteína 10 de la LLC-Linfoma de Células B/metabolismo , Neoplasias de la Mama/patología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , FN-kappa B/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Angiotensina/metabolismo , Animales , Apoptosis , Proteína 10 de la LLC-Linfoma de Células B/antagonistas & inhibidores , Proteína 10 de la LLC-Linfoma de Células B/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Proteínas Adaptadoras de Señalización CARD/genética , Movimiento Celular , Proliferación Celular , Embrión de Pollo , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/antagonistas & inhibidores , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , FN-kappa B/genética , Neovascularización Patológica , Pronóstico , ARN Interferente Pequeño/genética , Receptor de Angiotensina Tipo 1/genética , Receptores de Angiotensina/química , Receptores de Angiotensina/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Viral Immunol ; 30(5): 377-387, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28410454

RESUMEN

Marek's disease virus (MDV), a highly cell-associated oncogenic α-herpesvirus, is the causative agent of T cell lymphoma and neuropathic disease called Marek's disease. The skin is the only anatomical site where infectious enveloped cell-free virions are produced and shed into the environment. Studies have demonstrated that MDV infection induces immunological responses within the skin, including the release of cytokines and the recruitment of T lymphocytes. The host immune response, however, is not sufficient to block replication and shedding of the virus particles from the skin. In this study, we examined the gene expression profiling in the skin tissues of MDV-infected chickens to identify viral-induced alterations in the host gene expression pattern. To identify these genes in an unbiased and comprehensive manner, we performed RNA-seq on skin samples of MDV-infected chickens at 10, 20, and 30 days postinfection (dpi). We identified 820, 1,333, and 1,571 upregulated genes in the skin of MDV-infected chickens at 10, 20, and 30 dpi, respectively. In addition, we identified 461, 878, and 1,751 downregulated genes corresponding to the same time points, respectively. Analysis of the upregulated genes resulted in the identification of multiple gene ontology (GO) categories, with most falling under the host immune response. Searching these immune related GO categories, we identified six genes, gga-let-7d, interleukin 22 receptor subunit alpha 2, tumor necrosis factor receptor superfamily member 21, Proline-serine-threonine phosphatase-interacting protein 2, Suppressor of cytokine signaling (SOCS)1, and SOCS3, with known immunosuppressive properties that are upregulated in the skin of MDV-infected chickens.


Asunto(s)
Perfilación de la Expresión Génica , Herpesvirus Gallináceo 2/inmunología , Enfermedad de Marek/inmunología , Piel/inmunología , Animales , Pollos , Interacciones Huésped-Patógeno , Evasión Inmune , Enfermedad de Marek/patología , Análisis de Secuencia de ARN , Piel/patología , Factores de Tiempo
12.
Cell Rep ; 17(1): 221-232, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681433

RESUMEN

Microvascular endothelial cells maintain a tight barrier to prevent passage of plasma and circulating immune cells into the extravascular tissue compartment, yet endothelial cells respond rapidly to vasoactive substances, including thrombin, allowing transient paracellular permeability. This response is a cornerstone of acute inflammation, but the mechanisms responsible are still incompletely understood. Here, we demonstrate that thrombin triggers MALT1 to proteolytically cleave cylindromatosis (CYLD). Fragmentation of CYLD results in microtubule disruption and a cascade of events leading to endothelial cell retraction and an acute permeability response. This finding reveals an unexpected role for the MALT1 protease, which previously has been viewed mostly as a driver of pro-inflammatory NF-κB signaling in lymphocytes. Thus, MALT1 not only promotes immune cell activation but also acutely regulates endothelial cell biology, actions that together facilitate tissue inflammation. Pharmacologic inhibition of MALT1 may therefore have synergistic impact by targeting multiple disparate steps in the overall inflammatory response.


Asunto(s)
Caspasas/inmunología , Cisteína Endopeptidasas/inmunología , Células Endoteliales/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Proteínas de Neoplasias/inmunología , Trombina/farmacología , Animales , Transporte Biológico , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Caspasas/genética , Línea Celular , Cisteína Endopeptidasas/genética , Enzima Desubiquitinante CYLD , Células Endoteliales/citología , Células Endoteliales/inmunología , Regulación de la Expresión Génica , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Ratones , Ratones Transgénicos , Microtúbulos/ultraestructura , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , FN-kappa B/genética , FN-kappa B/inmunología , Proteínas de Neoplasias/genética , Permeabilidad/efectos de los fármacos , Cultivo Primario de Células , Receptor PAR-1/genética , Receptor PAR-1/inmunología , Transducción de Señal , Trombina/metabolismo
13.
J Biomol Screen ; 20(5): 673-80, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25550354

RESUMEN

Alphaviruses are a prominent class of reemergent pathogens due to their globally expanding ranges, potential for lethality, and possible use as bioweapons. The absence of effective treatments for alphaviruses highlights the need for innovative strategies to identify antiviral agents. Primary screens that use noninfectious self-replicating RNAs, termed replicons, have been used to identify potential antiviral compounds for alphaviruses. Only inhibitors of viral genome replication, however, will be identified using replicons, which excludes many other druggable steps in the viral life cycle. To address this limitation, we developed a western equine encephalitis virus pseudoinfectious particle system that reproduces several crucial viral life cycle steps in addition to genome replication. We used this system to screen a library containing ~26,000 extracts derived from marine microbes, and we identified multiple bacterial strains that produce compounds with potential antiviral activity. We subsequently used pseudoinfectious particle and replicon assays in parallel to counterscreen candidate extracts, and followed antiviral activity during biochemical fractionation and purification to differentiate between inhibitors of viral entry and genome replication. This novel process led to the isolation of a known alphavirus entry inhibitor, bafilomycin, thereby validating the approach for the screening and identification of potential antiviral compounds.


Asunto(s)
Alphavirus/efectos de los fármacos , Alphavirus/fisiología , Antivirales/farmacología , Productos Biológicos/farmacología , Descubrimiento de Drogas/métodos , Animales , Antivirales/química , Productos Biológicos/química , Línea Celular , Relación Dosis-Respuesta a Droga , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana/métodos , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas , Replicación Viral/efectos de los fármacos
14.
PLoS One ; 8(12): e82318, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24349254

RESUMEN

Natural products provide a vast array of chemical structures to explore in the discovery of new medicines. Although secondary metabolites produced by microbes have been developed to treat a variety of diseases, including bacterial and fungal infections, to date there has been limited investigation of natural products with antiviral activity. In this report, we used a phenotypic cell-based replicon assay coupled with an iterative biochemical fractionation process to identify, purify, and characterize antiviral compounds produced by marine microbes. We isolated a compound from Streptomyces kaviengensis, a novel actinomycetes isolated from marine sediments obtained off the coast of New Ireland, Papua New Guinea, which we identified as antimycin A1a. This compound displays potent activity against western equine encephalitis virus in cultured cells with half-maximal inhibitory concentrations of less than 4 nM and a selectivity index of greater than 550. Our efforts also revealed that several antimycin A analogues display antiviral activity, and mechanism of action studies confirmed that these Streptomyces-derived secondary metabolites function by inhibiting the cellular mitochondrial electron transport chain, thereby suppressing de novo pyrimidine synthesis. Furthermore, we found that antimycin A functions as a broad spectrum agent with activity against a wide range of RNA viruses in cultured cells, including members of the Togaviridae, Flaviviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Finally, we demonstrate that antimycin A reduces central nervous system viral titers, improves clinical disease severity, and enhances survival in mice given a lethal challenge with western equine encephalitis virus. Our results provide conclusive validation for using natural product resources derived from marine microbes as source material for antiviral drug discovery, and they indicate that host mitochondrial electron transport is a viable target for the continued development of broadly active antiviral compounds.


Asunto(s)
Actinobacteria/química , Antivirales/farmacología , Sedimentos Geológicos/microbiología , Animales , Antimicina A/química , Antimicina A/farmacología , Antimicina A/uso terapéutico , Antivirales/química , Antivirales/aislamiento & purificación , Productos Biológicos/química , Productos Biológicos/aislamiento & purificación , Productos Biológicos/farmacología , Línea Celular , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Fraccionamiento Químico , Transporte de Electrón/efectos de los fármacos , Virus de la Encefalitis/efectos de los fármacos , Encefalitis por Arbovirus/tratamiento farmacológico , Encefalitis por Arbovirus/patología , Encefalitis por Arbovirus/virología , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , ARN Viral/metabolismo , Estándares de Referencia , Reproducibilidad de los Resultados , Streptomyces/química , Análisis de Supervivencia , Transcripción Genética/efectos de los fármacos
15.
Cell Rep ; 1(5): 444-52, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22708078

RESUMEN

Excess serum free fatty acids (FFAs) are fundamental to the pathogenesis of insulin resistance. With high-fat feeding, FFAs activate NF-kB in target tissues, initiating negative crosstalk with insulin signaling. However, the mechanisms underlying FFA-dependent NF-kB activation remain unclear. Here, we demonstrate that the saturated FA, palmitate, requires Bcl10 for NF-kB activation in hepatocytes. Uptake of palmitate, metabolism to diacylglycerol, and subsequent activation of protein kinase C (PKC) appear to mechanistically link palmitate with Bcl10, known as a central component of a signaling complex that, along with CARMA3 and MALT1, activates NF-kB downstream of selected cell surface receptors. Consequently, Bcl10-deficient mice are protected from hepatic NF-kB activation and insulin resistance following brief high-fat diet, suggesting that Bcl10 plays a major role in the metabolic consequences of acute overnutrition. Surprisingly, while CARMA3 also participates in the palmitate response, MALT1 is completely dispensable, thereby revealing an apparent nonclassical role for Bcl10 in NF-kB signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma Hepatocelular/metabolismo , Ácidos Grasos/farmacología , Hepatocitos/metabolismo , Resistencia a la Insulina/fisiología , Neoplasias Hepáticas/metabolismo , FN-kappa B/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína 10 de la LLC-Linfoma de Células B , Proteínas Adaptadoras de Señalización CARD/metabolismo , Carcinoma Hepatocelular/patología , Caspasas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Dieta Alta en Grasa , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Proteínas de Neoplasias/metabolismo , Hipernutrición/metabolismo , Palmitatos/farmacología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA