Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Cell ; 159(7): 1615-25, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25525879

RESUMEN

Sirtuins (SIRTs) are critical enzymes that govern genome regulation, metabolism, and aging. Despite conserved deacetylase domains, mitochondrial SIRT4 and SIRT5 have little to no deacetylase activity, and a robust catalytic activity for SIRT4 has been elusive. Here, we establish SIRT4 as a cellular lipoamidase that regulates the pyruvate dehydrogenase complex (PDH). Importantly, SIRT4 catalytic efficiency for lipoyl- and biotinyl-lysine modifications is superior to its deacetylation activity. PDH, which converts pyruvate to acetyl-CoA, has been known to be primarily regulated by phosphorylation of its E1 component. We determine that SIRT4 enzymatically hydrolyzes the lipoamide cofactors from the E2 component dihydrolipoyllysine acetyltransferase (DLAT), diminishing PDH activity. We demonstrate SIRT4-mediated regulation of DLAT lipoyl levels and PDH activity in cells and in vivo, in mouse liver. Furthermore, metabolic flux switching via glutamine stimulation induces SIRT4 lipoamidase activity to inhibit PDH, highlighting SIRT4 as a guardian of cellular metabolism.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Sirtuinas/metabolismo , Amidohidrolasas/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Glutamina/metabolismo , Humanos , Hígado/metabolismo , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Ratas , Sirtuinas/genética , Ácido Tióctico/análogos & derivados , Ácido Tióctico/metabolismo
2.
Cell ; 151(7): 1569-80, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260143

RESUMEN

Many signaling circuits face a fundamental tradeoff between accelerating their response speed while maintaining final levels below a cytotoxic threshold. Here, we describe a transcriptional circuitry that dynamically converts signaling inputs into faster rates without amplifying final equilibrium levels. Using time-lapse microscopy, we find that transcriptional activators accelerate human cytomegalovirus (CMV) gene expression in single cells without amplifying steady-state expression levels, and this acceleration generates a significant replication advantage. We map the accelerator to a highly self-cooperative transcriptional negative-feedback loop (Hill coefficient ∼7) generated by homomultimerization of the virus's essential transactivator protein IE2 at nuclear PML bodies. Eliminating the IE2-accelerator circuit reduces transcriptional strength through mislocalization of incoming viral genomes away from PML bodies and carries a heavy fitness cost. In general, accelerators may provide a mechanism for signal-transduction circuits to respond quickly to external signals without increasing steady-state levels of potentially cytotoxic molecules.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/genética , Redes Reguladoras de Genes , Aptitud Genética , Citomegalovirus/fisiología , Retroalimentación Fisiológica , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Cuerpos de Inclusión Viral/metabolismo , Imagen de Lapso de Tiempo , Transactivadores/metabolismo , Activación Transcripcional , Replicación Viral
3.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33723080

RESUMEN

The tryptophan metabolite, kynurenine, is known to be produced at elevated levels within human cytomegalovirus (HCMV)-infected fibroblasts. Kynurenine is an endogenous aryl hydrocarbon receptor (AhR) ligand. Here we show that the AhR is activated following HCMV infection, and pharmacological inhibition of AhR or knockdown of AhR RNA reduced the accumulation of viral RNAs and infectious progeny. RNA-seq analysis of infected cells following AhR knockdown showed that the receptor alters the levels of numerous RNAs, including RNAs related to cell cycle progression. AhR knockdown alleviated the G1/S cell cycle block that is normally instituted in HCMV-infected fibroblasts, consistent with its known ability to regulate cell cycle progression and cell proliferation. In sum, AhR is activated by kynurenine and perhaps other ligands produced during HCMV infection, it profoundly alters the infected-cell transcriptome, and one outcome of its activity is a block to cell cycle progression, providing mechanistic insight to a long-known element of the virus-host cell interaction.


Asunto(s)
Ciclo Celular , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Fase G1 , Receptores de Hidrocarburo de Aril/metabolismo , División Celular , Infecciones por Citomegalovirus/genética , Interacciones Huésped-Patógeno , Humanos , Quinurenina/metabolismo , Ligandos
4.
Proc Natl Acad Sci U S A ; 117(50): 31891-31901, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33257565

RESUMEN

Pericentromeric human satellite II (HSATII) repeats are normally silent but can be actively transcribed in tumor cells, where increased HSATII copy number is associated with a poor prognosis in colon cancer, and in human cytomegalovirus (HCMV)-infected fibroblasts, where the RNA facilitates viral replication. Here, we report that HCMV infection or treatment of ARPE-19 diploid epithelial cells with DNA-damaging agents, etoposide or zeocin, induces HSATII RNA expression, and a kinase-independent function of ATM is required for the induction. Additionally, various breast cancer cell lines growing in adherent, two-dimensional cell culture express HSATII RNA at different levels, and levels are markedly increased when cells are infected with HCMV or treated with zeocin. High levels of HSATII RNA expression correlate with enhanced migration of breast cancer cells, and knockdown of HSATII RNA reduces cell migration and the rate of cell proliferation. Our investigation links high expression of HSATII RNA to the DNA damage response, centered on a noncanonical function of ATM, and demonstrates a role for the satellite RNA in tumor cell proliferation and movement.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Neoplasias de la Mama/genética , Infecciones por Citomegalovirus/genética , ADN Satélite/genética , ARN no Traducido/genética , Bleomicina/farmacología , Bleomicina/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/virología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Daño del ADN/efectos de los fármacos , Reparación del ADN , Progresión de la Enfermedad , Etopósido/farmacología , Etopósido/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Aptitud Genética/efectos de los fármacos , Humanos , ARN no Traducido/metabolismo , RNA-Seq , Secuencias Repetitivas de Ácidos Nucleicos , Activación Transcripcional/efectos de los fármacos
5.
Proc Natl Acad Sci U S A ; 116(38): 18971-18982, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31481624

RESUMEN

Human cytomegalovirus (HCMV) manipulates many aspects of host cell biology to create an intracellular milieu optimally supportive of its replication and spread. Our study reveals that levels of several components of the purinergic signaling system, including the P2Y2 and P2X5 receptors, are elevated in HCMV-infected fibroblasts. Knockdown and drug treatment experiments demonstrated that P2Y2 enhances the yield of virus, whereas P2X5 reduces HCMV production. The HCMV IE1 protein induces P2Y2 expression; and P2Y2-mediated signaling is important for efficient HCMV gene expression, DNA synthesis, and the production of infectious HCMV progeny. P2Y2 cooperates with the viral UL37x1 protein to regulate cystolic Ca2+ levels. P2Y2 also regulates PI3K/Akt signaling and infected cell motility. Thus, P2Y2 functions at multiple points within the viral replication cycle to support the efficient production of HCMV progeny, and it may facilitate in vivo viral spread through its role in cell migration.


Asunto(s)
Calcio/metabolismo , Movimiento Celular , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Receptores Purinérgicos P2Y2/metabolismo , Línea Celular , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/patología , ADN Viral/metabolismo , Fibroblastos/metabolismo , Fibroblastos/virología , Expresión Génica , Técnicas de Silenciamiento del Gen , Interacciones Huésped-Patógeno , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antagonistas del Receptor Purinérgico P2/farmacología , Receptores Purinérgicos P2X5/genética , Receptores Purinérgicos P2X5/metabolismo , Receptores Purinérgicos P2Y2/genética , Transducción de Señal , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
6.
Proc Natl Acad Sci U S A ; 115(42): E9889-E9898, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30275317

RESUMEN

Human CMV (HCMV) exhibits a broad cell tropism that depends on two virion glycoprotein complexes: a trimeric complex (gH/gL/gO) that facilitates viral infection primarily in fibroblasts and a pentameric complex (gH/gL/pUL128-pUL130-pUL131A) that mediates infection in epithelial and endothelial cells. We performed genome-wide CRISPR screens in which the PDGF receptor-α (PDGFRα) was identified as the most significant cellular gene product essential for infection by HCMV virions containing only trimeric complex (trimer-only virus). Trimer-only virus did not enter PDGFRα knockout fibroblasts. By using knockout fibroblasts, the extracellular domain of PDGFRα required for virus entry was mapped, and the intracellular tyrosine kinase domain was shown to be nonessential. In addition, direct cell-to-cell spread of virus from knockout cells transfected with trimer-only viral DNA was blocked, despite the production of infectious virus in the transfected cells. In contrast to trimer-only virus, wild-type HCMV virions containing both trimeric and pentameric complexes entered PDGFRα knockout cells, reinforcing the view that fibroblasts contain a second, independent receptor for the pentameric complex. Importantly, however, wild-type virus entered the knockout fibroblasts at reduced efficiency compared with parental fibroblasts, arguing that the cellular receptor for the virion pentameric complex is limiting or that virions are produced containing different relative amounts of the two glycoprotein complexes. Finally, ectopic expression of PDGFRα in ARPE-19 epithelial cells and THP-1 monocytic cells, which have little to no endogenous PDGFRα expression, markedly enhanced their susceptibility to trimer-only virions. In sum, our data clarify several key determinants of HCMV tropism.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Fibroblastos/virología , Pulmón/virología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Virión , Internalización del Virus , Células Cultivadas , Infecciones por Citomegalovirus/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Pulmón/citología , Pulmón/metabolismo , Proteínas del Envoltorio Viral/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(39): E8244-E8253, 2017 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-28874566

RESUMEN

Human cytomegalovirus (HCMV) is the prototypical human ß-herpes virus. Here we perform a systems analysis of the HCMV host-cell transcriptome, using gene set enrichment analysis (GSEA) as an engine to globally map the host-pathogen interaction across two cell types. Our analysis identified several previously unknown signatures of infection, such as induction of potassium channels and amino acid transporters, derepression of genes marked with histone H3 lysine 27 trimethylation (H3K27me3), and inhibition of genes related to epithelial-to-mesenchymal transition (EMT). The repression of EMT genes was dependent on early viral gene expression and correlated with induction E-cadherin (CDH1) and mesenchymal-to-epithelial transition (MET) genes. Infection of transformed breast carcinoma and glioma stem cells similarly inhibited EMT and induced MET, arguing that HCMV induces an epithelium-like cellular environment during infection.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Transición Epitelial-Mesenquimal/inmunología , Antígenos CD , Cadherinas/inmunología , Línea Celular Tumoral , Infecciones por Citomegalovirus/patología , Histonas/inmunología , Humanos
8.
Proc Natl Acad Sci U S A ; 111(12): E1140-8, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24616524

RESUMEN

The human cytomegalovirus immediate-early protein pUL37x1 induces the release of Ca(2+) stores from the endoplasmic reticulum into the cytosol. This release causes reorganization of the cellular actin cytoskeleton with concomitant cell rounding. Here we demonstrate that pUL37x1 activates Ca(2+)-dependent protein kinase Cα (PKCα). Both PKCα and Rho-associated protein kinases are required for actin reorganization and cell rounding; however, only PKCα is required for the efficient production of virus progeny, arguing that HCMV depends on the kinase for a second function. PKCα activation is also needed for the production of large (1-5 µm) cytoplasmic vesicles late after infection. The production of these vesicles is blocked by inhibition of fatty acid or phosphatidylinositol-3-phosphate biosynthesis, and the failure to produce vesicles is correlated with substantially reduced production of enveloped virus capsids. These results connect earlier work identifying a requirement for lipid synthesis with specific morphological changes, and support the argument that the PKCα-induced large vesicles are either required for the efficient production of mature virus particles or serve as a marker for the process.


Asunto(s)
Calcio/metabolismo , Forma de la Célula , Citoplasma/metabolismo , Proteínas Inmediatas-Precoces/fisiología , Proteína Quinasa C-alfa/metabolismo , Secuencia de Bases , Membrana Celular/enzimología , Citoplasma/enzimología , Activación Enzimática , Transporte Iónico , ARN Interferente Pequeño/genética
9.
Proc Natl Acad Sci U S A ; 111(52): E5706-15, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512541

RESUMEN

An shRNA-mediated screen of the 48 human nuclear receptor genes identified multiple candidates likely to influence the production of human cytomegalovirus in cultured human fibroblasts, including the estrogen-related receptor α (ERRα), an orphan nuclear receptor. The 50-kDa receptor and a 76-kDa variant were induced posttranscriptionally following infection. Genetic and pharmacological suppression of the receptor reduced viral RNA, protein, and DNA accumulation, as well as the yield of infectious progeny. In addition, RNAs encoding multiple metabolic enzymes, including enzymes sponsoring glycolysis (enolase 1, triosephosphate isomerase 1, and hexokinase 2), were reduced when the function of ERRα was inhibited in infected cells. Consistent with the effect on RNAs, a substantial number of metabolites, which are normally induced by infection, were either not increased or were increased to a reduced extent in the absence of normal ERRα activity. We conclude that ERRα is needed for the efficient production of cytomegalovirus progeny, and we propose that the nuclear receptor contributes importantly to the induction of a metabolic environment that supports optimal cytomegalovirus replication.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Receptor alfa de Estrógeno/metabolismo , Replicación Viral/fisiología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Glucólisis/genética , Hexoquinasa/genética , Hexoquinasa/metabolismo , Humanos , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Biosíntesis de Proteínas/genética , ARN Viral/genética , ARN Viral/metabolismo , Triosa-Fosfato Isomerasa/genética , Triosa-Fosfato Isomerasa/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Mol Cell Proteomics ; 13(9): 2399-410, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24987098

RESUMEN

This work represents the first comprehensive quantitative analysis of global histone post-translational modifications (PTMs) from a virus infection, namely human cytomegalovirus (HCMV) infection. We used a nanoLC-MS/MS platform to identify and quantify the dynamic histone H3 and H4 PTMs expressed during HCMV replication in primary fibroblasts. Specifically, we examined the changes in histone PTMs over a 96 h time course to sample the immediate early (IE), early (E), and late (L) stages of viral infection. Several changes in histone H3 and H4 PTMs were observed, including a marked increase in H3K79me2 and H3K27me3K36me2, and a decrease in H4K16ac, highlighting likely epigenetic strategies of transcriptional activation and silencing during HCMV lytic infection. Heavy methyl-SILAC (hm-SILAC) was used to further confirm the histone methylation flux (especially for H3K79) during HCMV infection. We evaluated DOT1L (the H3K79 methyltransferase) mRNA levels in mock and HCMV-infected cells over a 96 h time course, and observed a significant increase in this methyltransferase as early as 24 hpi showing that viral infection up-regulates DOT1L expression, which drives H3K79me2. We then used shRNA to create a DOT1L knockdown cell population, and found that HCMV infection of the knockdown cells resulted in a 10-fold growth defect when compared with infected control cells not subjected to knockdown. This work documents multiple histone PTMs that occur in response to HCMV infection of fibroblasts, and provides a framework for evaluation of the role of epigenetic modifications in the virus-host interaction.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Fibroblastos/virología , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Línea Celular , Cromatografía Liquida , Citomegalovirus/genética , Citomegalovirus/metabolismo , ADN Viral/análisis , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina , Humanos , Metiltransferasas/genética , Proteómica , Espectrometría de Masas en Tándem , Proteínas Virales/metabolismo , Replicación Viral/fisiología
11.
Proc Natl Acad Sci U S A ; 110(51): E5006-15, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297925

RESUMEN

Herpes simplex virus 1 (HSV-1) infection triggers specific metabolic changes in its host cell. To explore the interactions between cellular metabolism and HSV-1 infection, we performed an siRNA screen of cellular metabolic genes, measuring their effect on viral replication. The screen identified multiple enzymes predicted to influence HSV-1 replication, including argininosuccinate synthetase 1 (AS1), which consumes aspartate as part of de novo arginine synthesis. Knockdown of AS1 robustly enhanced viral genome replication and the production of infectious virus. Using high-resolution liquid chromatography-mass spectrometry, we found that the metabolic phenotype induced by knockdown of AS1 in human fibroblasts mimicked multiple aspects of the metabolic program observed during HSV-1 infection, including an increase in multiple nucleotides and their precursors. Together with the observation that AS1 protein and mRNA levels decrease during wild-type infection, this work suggests that reduced AS1 activity is partially responsible for the metabolic program induced by infection.


Asunto(s)
Argininosuccinato Sintasa/metabolismo , Fibroblastos/enzimología , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Replicación Viral/fisiología , Animales , Argininosuccinato Sintasa/genética , Chlorocebus aethiops , Fibroblastos/patología , Fibroblastos/virología , Técnicas de Silenciamiento del Gen , Genoma Viral/fisiología , Herpes Simple/genética , Herpes Simple/patología , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Vero
12.
Proteomics ; 15(12): 2006-22, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25867546

RESUMEN

Replication of human cytomegalovirus (HCMV) is regulated in part by cellular kinases and the single viral Ser/Thr kinase, pUL97. The virus-coded kinase augments the replication of HCMV by enabling nuclear egress and altering cell cycle progression. These roles are accomplished through direct phosphorylation of nuclear lamins and the retinoblastoma protein, respectively. In an effort to identify additional pUL97 substrates, we analyzed the phosphoproteome of SILAC-labeled human fibroblasts during infection with either wild-type HCMV or a pUL97 kinase-dead mutant virus. Phosphopeptides were enriched over a titanium dioxide matrix and analyzed by high-resolution MS. We identified 157 unambiguous phosphosites from 106 cellular and 17 viral proteins whose phosphorylation required UL97. Analysis of peptides containing these sites allowed the identification of several candidate pUL97 phosphorylation motifs, including a completely novel phosphorylation motif, LxSP. Substrates harboring the LxSP motif were enriched in nucleocytoplasmic transport functions, including a number of components of the nuclear pore complex. These results extend the known functions of pUL97 and suggest that modulation of nuclear pore function may be important during HCMV replication.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Fibroblastos/metabolismo , Fosfoproteínas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteoma/análisis , Proteómica/métodos , Western Blotting , Células Cultivadas , Cromatografía Liquida , Infecciones por Citomegalovirus/virología , Fibroblastos/virología , Humanos , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Espectrometría de Masas en Tándem , Replicación Viral
13.
Proteomics ; 15(12): 1983-94, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25894605

RESUMEN

Viruses rely on the host translation machinery for the synthesis of viral proteins. Human cells have evolved sensors that recognize viral RNAs and inhibit mRNA translation in order to limit virus replication. Understanding how viruses manipulate the host translation machinery to gain access to ribosomes and disable the antiviral response is therefore a critical aspect of the host/pathogen interface. In this study, we used a proteomics approach to identify human cytomegalovirus (HCMV) proteins that might contribute to viral mRNA translation. The HCMV TRS1 protein (pTRS1) associated with the 7-methylguanosine mRNA cap, increased the total level of protein synthesis, and colocalized with mRNAs undergoing translation initiation during infection. pTRS1 stimulated translation of a nonviral reporter gene and increased the translation of a reporter containing an HCMV 5' untranslated region (5'UTR) to a greater extent. The preferential effect of pTRS1 on translation of an mRNA containing a viral 5'UTR required the pTRS1 RNA and double-stranded RNA-dependent protein kinase (PKR)-binding domains, and was likely the result of PKR inhibition. However, pTRS1 also stimulated the total level of protein synthesis and translation directed by an HCMV 5'UTR in cells lacking PKR. Thus our results demonstrate that pTRS1 stimulates translation through both PKR-dependent and PKR-independent mechanisms.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Guanosina/análogos & derivados , Biosíntesis de Proteínas , Proteínas de Unión a Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Proteínas Virales/metabolismo , Western Blotting , Células Cultivadas , Infecciones por Citomegalovirus/virología , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Guanosina/química , Guanosina/metabolismo , Células HeLa , Humanos , Inmunoprecipitación , Proteínas de Unión a Caperuzas de ARN/genética , ARN Mensajero/genética , ARN Viral/genética , Ribosomas/metabolismo , Proteínas Virales/genética , Replicación Viral
14.
PLoS Pathog ; 9(5): e1003333, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696731

RESUMEN

Human cytomegalovirus hijacks host cell metabolism, increasing the flux of carbon from glucose to malonyl-CoA, the committed precursor to fatty acid synthesis and elongation. Inhibition of acetyl-CoA carboxylase blocks the production of progeny virus. To probe further the role of fatty acid metabolism during infection, we performed an siRNA screen to identify host cell metabolic enzymes needed for the production of infectious cytomegalovirus progeny. The screen predicted that multiple long chain acyl-CoA synthetases and fatty acid elongases are needed during infection, and the levels of RNAs encoding several of these enzymes were upregulated by the virus. Roles for acyl-CoA synthetases and elongases during infection were confirmed by using small molecule antagonists. Consistent with a role for these enzymes, mass spectrometry-based fatty acid analysis with ¹³C-labeling revealed that malonyl-CoA is consumed by elongases to produce very long chain fatty acids, generating an approximately 8-fold increase in C26-C34 fatty acid tails in infected cells. The virion envelope was yet further enriched in C26-C34 saturated fatty acids, and elongase inhibitors caused the production of virions with lower levels of these fatty acids and markedly reduced infectivity. These results reveal a dependence of cytomegalovirus on very long chain fatty acid metabolism.


Asunto(s)
Coenzima A Ligasas/metabolismo , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Ácidos Grasos/biosíntesis , Malonil Coenzima A/metabolismo , Virión/metabolismo , Línea Celular , Coenzima A Ligasas/genética , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Ácidos Grasos/genética , Humanos , Malonil Coenzima A/genética , ARN Interferente Pequeño , Virión/genética
15.
Proc Natl Acad Sci U S A ; 109(8): 3071-6, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22315427

RESUMEN

Human cytomegalovirus (HCMV) modulates numerous cellular signaling pathways. Alterations in signaling are evident from the broad changes in cellular phosphorylation that occur during HCMV infection and from the altered activity of multiple kinases. Here we report a comprehensive RNAi screen, which predicts that 106 cellular kinases influence growth of the virus, most of which were not previously linked to HCMV replication. Multiple elements of the AMP-activated protein kinase (AMPK) pathway scored in the screen. As a regulator of carbon and nucleotide metabolism, AMPK is poised to activate many of the metabolic pathways induced by HCMV infection. An AMPK inhibitor, compound C, blocked a substantial portion of HCMV-induced metabolic changes, inhibited the accumulation of all HCMV proteins tested, and markedly reduced the production of infectious progeny. We propose that HCMV requires AMPK or related activity for viral replication and reprogramming of cellular metabolism.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Infecciones por Citomegalovirus/enzimología , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Proteómica/métodos , Línea Celular , Citomegalovirus/efectos de los fármacos , Humanos , Metaboloma/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 109(24): 9575-80, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22645331

RESUMEN

Cell proteins can restrict the replication of viruses. Here, we identify the cellular BclAF1 protein as a human cytomegalovirus restriction factor and describe two independent mechanisms the virus uses to decrease its steady-state levels. Immediately following infection, the viral pp71 and UL35 proteins, which are delivered to cells within virions, direct the proteasomal degradation of BclAF1. Although BclAF1 reaccumulates through the middle stages of infection, it is subsequently down-regulated at late times by miR-UL112-1, a virus-encoded microRNA. In the absence of BclAF1 neutralization, viral gene expression and replication are inhibited. These data identify two temporally and mechanistically distinct functions used by human cytomegalovirus to down-regulate a cellular antiviral protein.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , MicroARNs/antagonistas & inhibidores , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Represoras/inmunología , Proteínas Supresoras de Tumor/inmunología , Citomegalovirus/genética , Genes Inmediatos-Precoces , Humanos , Hidrólisis , MicroARNs/metabolismo
17.
Proc Natl Acad Sci U S A ; 108(31): 12869-74, 2011 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-21768361

RESUMEN

Human cytomegalovirus induces and requires fatty acid synthesis. This suggests an essential role for lipidome remodeling in viral replication. We used mass spectrometry to quantify glycerophospholipids in mock-infected and virus-infected fibroblasts, as well as in virions. Although the lipid composition of mock-infected and virus-infected fibroblasts was similar, virions were markedly different. The virion envelope contained twofold more phosphatidylethanolamines and threefold less phosphatidylserines than the host cell. This indicates that the virus buds from a membrane with a different lipid composition from the host cell as a whole. Compared with published datasets, the virion envelope showed the greatest similarity to the synaptic vesicle lipidome. Synaptosome-associated protein of 25 kDa (SNAP-25) is a component of the complex that mediates exocytosis of synaptic vesicles in neurons; and its homolog, SNAP-23, functions in exocytosis in many other cell types. Infection induced the relocation of SNAP-23 to the cytoplasmic viral assembly zone, and knockdown of SNAP-23 inhibited the production of virus. We propose that cytomegalovirus capsids acquire their envelope by budding into vesicles with a lipid composition similar to that of synaptic vesicles, which subsequently fuse with the plasma membrane to release virions from the cell.


Asunto(s)
Citomegalovirus/química , Lípidos/química , Proteínas SNARE/metabolismo , Virión/química , Western Blotting , Línea Celular , Células Cultivadas , Cromatografía Liquida , Citomegalovirus/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/virología , Técnica del Anticuerpo Fluorescente , Glicerofosfolípidos/química , Glicerofosfolípidos/metabolismo , Interacciones Huésped-Patógeno , Humanos , Masculino , Espectrometría de Masas , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Proteínas Qb-SNARE/genética , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/genética , Proteínas Qc-SNARE/metabolismo , Interferencia de ARN , Proteínas SNARE/genética , Vesículas Sinápticas/química , Virión/fisiología , Replicación Viral
18.
Antiviral Res ; 226: 105888, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38641024

RESUMEN

296 million people worldwide are predisposed to developing severe end-stage liver diseases due to chronic hepatitis B virus (HBV) infection. HBV forms covalently closed circular DNA (cccDNA) molecules that persist as episomal DNA in the nucleus of infected hepatocytes and drive viral replication. Occasionally, the HBV genome becomes integrated into host chromosomal DNA, a process that is believed to significantly contribute to circulating HBsAg levels and HCC development. Neither cccDNA accumulation nor expression from integrated HBV DNA are directly targeted by current antiviral treatments. In this study, we investigated the antiviral properties of a newly described allosteric modulator, FLS-359, that targets sirtuin 2 (SIRT2), an NAD+-dependent deacylase. Our results demonstrate that SIRT2 modulation by FLS-359 and by other tool compounds inhibits cccDNA synthesis following de novo infection of primary human hepatocytes and HepG2 (C3A)-NTCP cells, and FLS-359 substantially reduces cccDNA recycling in HepAD38 cells. While pre-existing cccDNA is not eradicated by short-term treatment with FLS-359, its transcriptional activity is substantially impaired, likely through inhibition of viral promoter activities. Consistent with the inhibition of viral transcription, HBsAg production by HepG2.2.15 cells, which contain integrated HBV genomes, is also suppressed by FLS-359. Our study provides further insights on SIRT2 regulation of HBV infection and supports the development of potent SIRT2 inhibitors as HBV antivirals.


Asunto(s)
Antivirales , ADN Circular , ADN Viral , Virus de la Hepatitis B , Hepatocitos , Sirtuina 2 , Replicación Viral , Humanos , ADN Circular/metabolismo , Sirtuina 2/antagonistas & inhibidores , Sirtuina 2/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Hepatocitos/virología , Hepatocitos/efectos de los fármacos , Antivirales/farmacología , Replicación Viral/efectos de los fármacos , Células Hep G2 , Regulación Alostérica/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
19.
J Virol ; 86(21): 11425-33, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22915800

RESUMEN

Human cytomegalovirus (HCMV) encodes four putative G protein-coupled receptors, including pUL78, whose rodent orthologues are known to be important for replication and spread in their hosts. To investigate the mechanism by which pUL78 contributes to viral replication and pathogenesis, we generated a derivative of the TB40/E clinical isolate of HCMV that is unable to express the receptor. Consistent with previous findings using laboratory strains of the virus, the mutant replicated normally in fibroblasts. Although laboratory strains are restricted to growth in fibroblasts, clinical isolates grow in many cell types, including epithelial and endothelial cells, in which the pUL78-deficient TB40/E derivative exhibited a growth defect. Infection with the mutant virus resulted in a significant decrease in viral RNA and protein expression. Although there was no difference in binding of the virus to the cell, we detected a delay in the entry and subsequent delivery of virion DNA and protein to the nuclei of epithelial cells following infection with the UL78 mutant virus. Taken together, our results demonstrate that pUL78 supports infection at a point after binding but before entry in epithelial cells, a cell type important for in vivo viral replication and spread.


Asunto(s)
Citomegalovirus/patogenicidad , Células Epiteliales/virología , Fibroblastos/virología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/fisiología , Humanos , Replicación Viral
20.
J Virol ; 86(15): 8259-68, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623791

RESUMEN

Herpes simplex virus 1 infection triggers multiple changes in the metabolism of host cells, including a dramatic decrease in the levels of NAD(+). In addition to its role as a cofactor in reduction-oxidation reactions, NAD(+) is required for certain posttranslational modifications. Members of the poly(ADP-ribose) polymerase (PARP) family of enzymes are major consumers of NAD(+), which they utilize to form poly(ADP-ribose) (PAR) chains on protein substrates in response to DNA damage. PAR chains can subsequently be removed by the enzyme poly(ADP-ribose) glycohydrolase (PARG). We report here that the HSV-1 infection-induced drop in NAD(+) levels required viral DNA replication, was associated with an increase in protein poly(ADP-ribosyl)ation (PARylation), and was blocked by pharmacological inhibition of PARP-1/PARP-2 (PARP-1/2). Neither virus yield nor the cellular metabolic reprogramming observed during HSV-1 infection was altered by the rescue or further depletion of NAD(+) levels. Expression of the viral protein ICP0, which possesses E3 ubiquitin ligase activity, was both necessary and sufficient for the degradation of the 111-kDa PARG isoform. This work demonstrates that HSV-1 infection results in changes to NAD(+) metabolism by PARP-1/2 and PARG, and as PAR chain accumulation can induce caspase-independent apoptosis, we speculate that the decrease in PARG levels enhances the auto-PARylation-mediated inhibition of PARP, thereby avoiding premature death of the infected cell.


Asunto(s)
Glicósido Hidrolasas/metabolismo , Herpes Simple/enzimología , Herpesvirus Humano 1/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteolisis , Células Cultivadas , Replicación del ADN/genética , ADN Viral/genética , ADN Viral/metabolismo , Glicósido Hidrolasas/genética , Herpes Simple/genética , Herpesvirus Humano 1/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , NAD/genética , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA