Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(14): 3774-3793.e25, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34115982

RESUMEN

Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.


Asunto(s)
Citomegalovirus/fisiología , Macrófagos Alveolares/virología , Animales , Presentación de Antígeno , Efecto Espectador , Ciclo Celular , Línea Celular Transformada , Reprogramación Celular , Citomegalovirus/patogenicidad , Citomegalovirus/ultraestructura , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Proteínas Fluorescentes Verdes/metabolismo , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/ultraestructura , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Células Madre/patología , Replicación Viral/fisiología , Vía de Señalización Wnt
2.
Traffic ; 20(2): 152-167, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30548142

RESUMEN

Morphogenesis of herpesviral virions is initiated in the nucleus but completed in the cytoplasm. Mature virions contain more than 25 tegument proteins many of which perform both nuclear and cytoplasmic functions suggesting they shuttle between these compartments. While nuclear import of herpesviral proteins was shown to be crucial for viral propagation, active nuclear export and its functional impact are still poorly understood. To systematically analyze nuclear export of tegument proteins present in virions of Herpes simplex virus type 1 (HSV1) and Epstein-Barr virus (EBV), the Nuclear EXport Trapped by RAPamycin (NEX-TRAP) was applied. Nine of the 22 investigated HSV1 tegument proteins including pUL4, pUL7, pUL11, pUL13, pUL21, pUL37d11, pUL47, pUL48 and pUS2 as well as 2 out of 6 EBV orthologs harbor nuclear export activity. A functional leucine-rich nuclear export sequence (NES) recognized by the export factor CRM1/Xpo1 was identified in six of them. The comparison between experimental and bioinformatic data indicates that experimental validation of predicted NESs is required. Mutational analysis of the pUL48/VP16 NES revealed its importance for herpesviral propagation. Together our data suggest that nuclear export is an important feature of the herpesviral life cycle required to co-ordinate nuclear and cytoplasmic processes.


Asunto(s)
Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 4/metabolismo , Señales de Exportación Nuclear , Proteínas de la Matriz Viral/química , Animales , Chlorocebus aethiops , Células HeLa , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 4/fisiología , Humanos , Células Vero , Proteínas de la Matriz Viral/metabolismo , Replicación Viral
3.
Plasmid ; 111: 102531, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32920019

RESUMEN

Engineering bacterial genomes or foreign DNA cloned as bacterial artificial chromosomes (BACs) relies on usage of helper plasmids, which deliver the desired tools transiently into the bacteria to be modified. After the anticipated action is completed the helper plasmids need to be cured. To make this efficient, plasmids are used that are maintained by conditional amplicons or carry a counter-selection marker. Here, we describe new conditional plasmids that can be maintained or cured by using chemical induction or repression. Our method is based on the dependency of plasmids carrying ori6Kγ origin of replication on the presence of protein Π. Ori6Kγ based plasmids are tightly regulated conditional constructs, but they require usually special E. coli strains to operate. To avoid this, we placed the Π protein expression under the control of a co-expressed conditional repressor. Regulating the maintenance of plasmids with administration or removal of chemicals is fully compatible with any other conditional amplicons applied to date. Here, we describe methods for inducing sites specific recombination of BACs as an example. However, the same strategy might be used to construct appropriate helper plasmids for any other transient components of genome editing methodologies such as λred recombinases or CRISPR/Cas components.


Asunto(s)
Escherichia coli/genética , Ingeniería Genética , Plásmidos/genética , Cromosomas Artificiales Bacterianos , Cromosomas Bacterianos , Replicación del ADN , Edición Génica , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Recombinación Genética , Temperatura
4.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29950411

RESUMEN

Herpesvirus infections are highly prevalent in the human population and persist for life. They are often acquired subclinically but potentially progress to life-threatening diseases in immunocompromised individuals. The interferon system is indispensable for the control of herpesviral replication. However, the responsible antiviral effector mechanisms are not well characterized. The type I interferon-induced, human myxovirus resistance 2 (MX2) gene product MxB, a dynamin-like large GTPase, has recently been identified as a potent inhibitor of HIV-1. We now show that MxB also interferes with an early step of herpesvirus replication, affecting alpha-, beta-, and gammaherpesviruses before or at the time of immediate early gene expression. Defined MxB mutants influencing GTP binding and hydrolysis revealed that the effector mechanism against herpesviruses is thoroughly different from that against HIV-1. Overall, our findings demonstrate that MxB serves as a broadly acting intracellular restriction factor that controls the establishment of not only retrovirus but also herpesvirus infection of all three subfamilies.IMPORTANCE Human herpesviruses pose a constant threat to human health. Reactivation of persisting herpesvirus infections, particularly in immunocompromised individuals and the elderly, can cause severe diseases, such as zoster, pneumonia, encephalitis, or cancer. The interferon system is relevant for the control of herpesvirus replication as exemplified by fatal disease outcomes in patients with primary immunodeficiencies. Here, we describe the interferon-induced, human MX2 gene product MxB as an efficient restriction factor of alpha-, beta-, and gammaherpesviruses. MxB has previously been described as an inhibitor of HIV-1. Importantly, our mutational analyses of MxB reveal an antiviral mechanism of herpesvirus restriction distinct from that against HIV-1. Thus, the dynamin-like MxB GTPase serves as a broadly acting intracellular restriction factor that controls retrovirus as well as herpesvirus infections.


Asunto(s)
Infecciones por Herpesviridae/prevención & control , Herpesviridae/fisiología , Mutación , Proteínas de Resistencia a Mixovirus/genética , Replicación Viral/genética , Células A549 , Herpesviridae/genética , Infecciones por Herpesviridae/virología , Humanos , Inmunidad Innata , Interferones , Proteínas de Resistencia a Mixovirus/inmunología , Replicación Viral/inmunología
5.
PLoS Pathog ; 12(12): e1006072, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27977791

RESUMEN

Cytomegalovirus (CMV) elicits long-term T-cell immunity of unparalleled strength, which has allowed the development of highly protective CMV-based vaccine vectors. Counterintuitively, experimental vaccines encoding a single MHC-I restricted epitope offered better immune protection than those expressing entire proteins, including the same epitope. To clarify this conundrum, we generated recombinant murine CMVs (MCMVs) encoding well-characterized MHC-I epitopes at different positions within viral genes and observed strong immune responses and protection against viruses and tumor growth when the epitopes were expressed at the protein C-terminus. We used the M45-encoded conventional epitope HGIRNASFI to dissect this phenomenon at the molecular level. A recombinant MCMV expressing HGIRNASFI on the C-terminus of M45, in contrast to wild-type MCMV, enabled peptide processing by the constitutive proteasome, direct antigen presentation, and an inflation of antigen-specific effector memory cells. Consequently, our results indicate that constitutive proteasome processing of antigenic epitopes in latently infected cells is required for robust inflationary responses. This insight allows utilizing the epitope positioning in the design of CMV-based vectors as a novel strategy for enhancing their efficacy.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Herpesviridae/inmunología , Epítopos Inmunodominantes/inmunología , Vacunas Virales/inmunología , Animales , Antígenos Virales/metabolismo , Cromatografía Liquida , Epítopos de Linfocito T/inmunología , Citometría de Flujo , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Espectrometría de Masas , Ratones , Muromegalovirus/inmunología , Mutagénesis Sitio-Dirigida , Péptidos , Vacunas Sintéticas/inmunología , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
6.
Virus Genes ; 53(5): 741-748, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28634751

RESUMEN

Herpesviruses are enveloped DNA viruses that infect vertebrate cells. Their high potential cloning capacity and the lifelong persistence of their genomes in various host cells make them attractive platforms for vector-based therapy. In this review, we would like to highlight recent advances of three major areas of herpesvirus vector development and application: (i) oncolytic therapy, (ii) recombinant vaccines, and (iii) large capacity gene transfer vehicles.


Asunto(s)
Vectores Genéticos/genética , Vectores Genéticos/inmunología , Herpesviridae/genética , Herpesviridae/inmunología , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Animales , Terapia Genética/métodos , Humanos , Vacunación/métodos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
7.
Nucleic Acids Res ; 43(8): e50, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25609697

RESUMEN

Recombinant adenoviruses containing a double-stranded DNA genome of 26-45 kb were broadly explored in basic virology, for vaccination purposes, for treatment of tumors based on oncolytic virotherapy, or simply as a tool for efficient gene transfer. However, the majority of recombinant adenoviral vectors (AdVs) is based on a small fraction of adenovirus types and their genetic modification. Recombineering techniques provide powerful tools for arbitrary engineering of recombinant DNA. Here, we adopted a seamless recombineering technology for high-throughput and arbitrary genetic engineering of recombinant adenoviral DNA molecules. Our cloning platform which also includes a novel recombination pipeline is based on bacterial artificial chromosomes (BACs). It enables generation of novel recombinant adenoviruses from different sources and switching between commonly used early generation AdVs and the last generation high-capacity AdVs lacking all viral coding sequences making them attractive candidates for clinical use. In combination with a novel recombination pipeline allowing cloning of AdVs containing large and complex transgenes and the possibility to generate arbitrary chimeric capsid-modified adenoviruses, these techniques allow generation of tailored AdVs with distinct features. Our technologies will pave the way toward broader applications of AdVs in molecular medicine including gene therapy and vaccination studies.


Asunto(s)
Adenoviridae/genética , Clonación Molecular/métodos , Ingeniería Genética , Vectores Genéticos , Recombinación Genética , Cromosomas Artificiales Bacterianos , ADN Viral , Células HEK293 , Humanos
8.
J Virol ; 89(4): 2121-35, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25473051

RESUMEN

UNLABELLED: Nuclear delivery of the adenoviral genome requires that the capsid cross the limiting membrane of the endocytic compartment and traverse the cytosol to reach the nucleus. This endosomal escape is initiated upon internalization and involves a highly coordinated process of partial disassembly of the entering capsid to release the membrane lytic internal capsid protein VI. Using wild-type and protein VI-mutated human adenovirus serotype 5 (HAdV-C5), we show that capsid stability and membrane rupture are major determinants of entry-related sorting of incoming adenovirus virions. Furthermore, by using electron cryomicroscopy, as well as penton- and protein VI-specific antibodies, we show that the amphipathic helix of protein VI contributes to capsid stability by preventing premature disassembly and deployment of pentons and protein VI. Thus, the helix has a dual function in maintaining the metastable state of the capsid by preventing premature disassembly and mediating efficient membrane lysis to evade lysosomal targeting. Based on these findings and structural data from cryo-electron microscopy, we suggest a refined disassembly mechanism upon entry. IMPORTANCE: In this study, we show the intricate connection of adenovirus particle stability and the entry-dependent release of the membrane-lytic capsid protein VI required for endosomal escape. We show that the amphipathic helix of the adenovirus internal protein VI is required to stabilize pentons in the particle while coinciding with penton release upon entry and that release of protein VI mediates membrane lysis, thereby preventing lysosomal sorting. We suggest that this dual functionality of protein VI ensures an optimal disassembly process by balancing the metastable state of the mature adenovirus particle.


Asunto(s)
Adenovirus Humanos/fisiología , Proteínas de la Cápside/metabolismo , Internalización del Virus , Desencapsidación Viral , Adenovirus Humanos/genética , Proteínas de la Cápside/genética , Línea Celular , Microscopía por Crioelectrón , Humanos
9.
J Immunol ; 193(5): 2306-16, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25070842

RESUMEN

Tuberculosis remains a global health problem so that a more effective vaccine than bacillus Calmette-Guérin is urgently needed. Cytomegaloviruses persist lifelong in vivo and induce powerful immune and increasing ("inflationary") responses, making them attractive vaccine vectors. We have used an m1-m16-deleted recombinant murine CMV (MCMV) expressing Mycobacterium tuberculosis Ag 85A to show that infection of mice with this recombinant significantly reduces the mycobacterial load after challenge with M. tuberculosis, whereas control empty virus has a lesser effect. Both viruses induce immune responses to H-2(d)-restricted epitopes of MCMV pp89 and M18 Ags characteristic of infection with other MCMVs. A low frequency of 85A-specific memory cells could be revealed by in vivo or in vitro boosting or after challenge with M. tuberculosis. Kinetic analysis of M. tuberculosis growth in the lungs of CMV-infected mice shows early inhibition of M. tuberculosis growth abolished by treatment with NK-depleting anti-asialo ganglio-N-tetraosylceramide Ab. Microarray analysis of the lungs of naive and CMV-infected mice shows increased IL-21 mRNA in infected mice, whereas in vitro NK assays indicate increased levels of NK activity. These data indicate that activation of NK cells by MCMV provides early nonspecific protection against M. tuberculosis, potentiated by a weak 85A-specific T cell response, and they reinforce the view that the innate immune system plays an important role in both natural and vaccine-induced protection against M. tuberculosis.


Asunto(s)
Epítopos/inmunología , Vectores Genéticos , Muromegalovirus , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/prevención & control , Animales , Epítopos/genética , Femenino , Antígeno de Histocompatibilidad H-2D/genética , Antígeno de Histocompatibilidad H-2D/inmunología , Interleucinas/genética , Interleucinas/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/genética , Vacunas contra la Tuberculosis/genética , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología
10.
Proc Natl Acad Sci U S A ; 110(24): E2191-8, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23708119

RESUMEN

Macrophages are diverse cell types in the first line of antimicrobial defense. Only a limited number of primary mouse models exist to study their function. Bone marrow-derived, macrophage-CSF-induced cells with a limited life span are the most common source. We report here a simple method yielding self-renewing, nontransformed, GM-CSF/signal transducer and activator of transcription 5-dependent macrophages (Max Planck Institute cells) from mouse fetal liver, which reflect the innate immune characteristics of alveolar macrophages. Max Planck Institute cells are exquisitely sensitive to selected microbial agents, including bacterial LPS, lipopeptide, Mycobacterium tuberculosis, cord factor, and adenovirus and mount highly proinflammatory but no anti-inflammatory IL-10 responses. They show a unique pattern of innate responses not yet observed in other mononuclear phagocytes. This includes differential LPS sensing and an unprecedented regulation of IL-1α production upon LPS exposure, which likely plays a key role in lung inflammation in vivo. In conclusion, Max Planck Institute cells offer an useful tool to study macrophage biology and for biomedical science.


Asunto(s)
Células de la Médula Ósea/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Macrófagos/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Interleucina-1alfa/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mycobacterium tuberculosis/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fagocitosis/inmunología , Propionibacterium acnes/inmunología , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Factor de Transcripción STAT5/metabolismo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Transcriptoma/inmunología
11.
J Gen Virol ; 96(11): 3313-3325, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26265177

RESUMEN

Herpes simplex virus type 1 (HSV-1) glycoprotein M (gM/UL10) is a 473 aa type III transmembrane protein that resides in various membrane compartments. HSV-1 gM contains several putative trafficking motifs, but their functional relevance remains to be elucidated. We show here that transiently expressed gM 19­343 was sufficient for transport to the trans-Golgi network (TGN), whilst gM 133­473, where the first two transmembrane domains were deleted, and gM 1­342, which lacked the final residue of the last transmembrane domain, were retained in the endoplasmic reticulum (ER), indicating that all transmembrane domains are required for proper folding and ER exit. A series of bacterial artificial chromosome mutants revealed that in addition to the authentic start codon, translation of gM can be initiated at methionine 19 and 133/135. Whilst a protein lacking the first 18 residues supported WT-like growth, gM 133/135­473 resulted in reduced plaque diameters resembling a UL10 deletion mutant. An HSV-1 mutant encoding gM 1­342 showed similar growth characteristics and accumulated non-enveloped cytoplasmic particles, whilst gM 1­343 resulted in a gain of function, indicating that all transmembrane domains of the protein are important for viral growth. A C-terminal extension further supported viral propagation; however, the C-terminal trafficking motifs (residues 423­473) were completely dispensable. We propose a functional core within gM 19­343 comprised of all transmembrane domains that is sufficient to target the protein to the TGN, a favoured site for envelopment, and to support viral functions.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Red trans-Golgi/virología , Secuencias de Aminoácidos , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , Glicoproteínas de Membrana/genética , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Virales/genética
12.
Genome Res ; 22(10): 2031-42, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22539649

RESUMEN

RNA synthesis and decay rates determine the steady-state levels of cellular RNAs. Metabolic tagging of newly transcribed RNA by 4-thiouridine (4sU) can reveal the relative contributions of RNA synthesis and decay rates. The kinetics of RNA processing, however, had so far remained unresolved. Here, we show that ultrashort 4sU-tagging not only provides snapshot pictures of eukaryotic gene expression but, when combined with progressive 4sU-tagging and RNA-seq, reveals global RNA processing kinetics at nucleotide resolution. Using this method, we identified classes of rapidly and slowly spliced/degraded introns. Interestingly, each class of splicing kinetics was characterized by a distinct association with intron length, gene length, and splice site strength. For a large group of introns, we also observed long lasting retention in the primary transcript, but efficient secondary splicing or degradation at later time points. Finally, we show that processing of most, but not all small nucleolar (sno)RNA-containing introns is remarkably inefficient with the majority of introns being spliced and degraded rather than processed into mature snoRNAs. In summary, our study yields unparalleled insights into the kinetics of RNA processing and provides the tools to study molecular mechanisms of RNA processing and their contribution to the regulation of gene expression.


Asunto(s)
Empalme del ARN , ARN/genética , ARN/metabolismo , Empalme Alternativo , Linfocitos B/metabolismo , Línea Celular , Exones , Humanos , Intrones , Cinética , ARN/química , Precursores del ARN/genética , Precursores del ARN/metabolismo , Sitios de Empalme de ARN , Estabilidad del ARN , Tiouridina/química , Transcripción Genética
13.
PLoS Pathog ; 9(8): e1003544, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950715

RESUMEN

During viral infection, a massive demand for viral glycoproteins can overwhelm the capacity of the protein folding and quality control machinery, leading to an accumulation of unfolded proteins in the endoplasmic reticulum (ER). To restore ER homeostasis, cells initiate the unfolded protein response (UPR) by activating three ER-to-nucleus signaling pathways, of which the inositol-requiring enzyme 1 (IRE1)-dependent pathway is the most conserved. To reduce ER stress, the UPR decreases protein synthesis, increases degradation of unfolded proteins, and upregulates chaperone expression to enhance protein folding. Cytomegaloviruses, as other viral pathogens, modulate the UPR to their own advantage. However, the molecular mechanisms and the viral proteins responsible for UPR modulation remained to be identified. In this study, we investigated the modulation of IRE1 signaling by murine cytomegalovirus (MCMV) and found that IRE1-mediated mRNA splicing and expression of the X-box binding protein 1 (XBP1) is repressed in infected cells. By affinity purification, we identified the viral M50 protein as an IRE1-interacting protein. M50 expression in transfected or MCMV-infected cells induced a substantial downregulation of IRE1 protein levels. The N-terminal conserved region of M50 was found to be required for interaction with and downregulation of IRE1. Moreover, UL50, the human cytomegalovirus (HCMV) homolog of M50, affected IRE1 in the same way. Thus we concluded that IRE1 downregulation represents a previously undescribed viral strategy to curb the UPR.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Endorribonucleasas/biosíntesis , Proteínas de la Membrana/biosíntesis , Muromegalovirus/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Respuesta de Proteína Desplegada , Animales , Línea Celular Transformada , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Endorribonucleasas/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Muromegalovirus/genética , Células 3T3 NIH , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/metabolismo , Proteína 1 de Unión a la X-Box
14.
J Immunol ; 190(7): 3399-409, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23460738

RESUMEN

Natural immunity to CMV dominates the CD4 and CD8 memory compartments of the CMV-seropositive host. This property has been recently exploited for experimental CMV-based vaccine vector strategies, and it has shown promise in animal models of AIDS and Ebola disease. Although it is generally agreed that CMV-based vaccine vectors may induce highly protective and persistent memory T cells, the influence of the gene expression context on Ag-specific T cell memory responses and immune protection induced by CMV vectors is not known. Using murine CMV (MCMV) recombinants expressing a single CD8 T cell epitope from HSV-1 fused to different MCMV genes, we show that magnitude and kinetics of T cell responses induced by CMV are dependent on the gene expression of CMV Ags. Interestingly, the kinetics of the immune response to the HSV-1 epitope was paralleled by a reciprocal depression of immune responses to endogenous MCMV Ags. Infection with a recombinant MCMV inducing a vigorous initial immune response to the recombinant peptide resulted in a depressed early response to endogenous MCMV Ag. Another recombinant virus, which induced a slowly developing "inflationary" T cell response to the HSV-1 peptide, induced weaker long-term responses to endogenous CMV Ags. Importantly, both mutants were able to protect mice from a challenge with HSV-1, mediating strong sterilizing immunity. Our data suggest that the context of gene expression markedly influences the T cell immunodominance hierarchy of CMV Ags, but the immune protection against HSV-1 does not require inflationary CD8 responses against the recombinant CMV-expressed epitope.


Asunto(s)
Antígenos Virales/genética , Antígenos Virales/inmunología , Expresión Génica , Epítopos Inmunodominantes/genética , Epítopos Inmunodominantes/inmunología , Muromegalovirus/genética , Muromegalovirus/inmunología , Animales , Antígenos Virales/química , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Orden Génico , Genoma Viral , Herpes Simple/inmunología , Herpes Simple/prevención & control , Herpesvirus Humano 1/inmunología , Masculino , Ratones , Mutación , Péptidos/química , Péptidos/genética , Péptidos/inmunología , Fenotipo , Replicación Viral
15.
J Virol ; 87(8): 4596-608, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23408606

RESUMEN

Human gammaherpesviruses cause morbidity and mortality associated with infection and transformation of lymphoid and endothelial cells. Knowledge of cell types involved in virus dissemination from primary virus entry to virus latency is fundamental for the understanding of gammaherpesvirus pathogenesis. However, the inability to directly trace cell types with respect to virus dissemination pathways has prevented definitive conclusions regarding the relative contribution of individual cell types. Here, we describe that the route of infection affects gammaherpesvirus dissemination pathways. We constructed a recombinant murine gammaherpesvirus 68 (MHV-68) variant harboring a cassette which switches fluorescent markers in a Cre-dependent manner. Since the recombinant virus which was constructed on the wild-type background was attenuated, in this study we used an M1-deleted version, which infected mice with normal kinetics. Infection of Cre-transgenic mice with this convertible virus was used to estimate the quantitative contribution of defined cell types to virus productivity and dissemination during the acute phase of MHV-68 infection. In systemic infection, we found splenic vascular endothelial cells (EC) among the first and main cells to produce virus. After local infection, the contribution of EC to splenic virus production did not represent such early kinetics. However, at later time points, B cell-derived viruses dominated splenic productivity independently of systemic or local infection. Systemic versus local infection also governed the cell types involved in loading peritoneal exudate cells, leading to latency in F4/80- and CD11b-positive target cells. Systemic infection supported EC-driven dissemination, whereas local infection supported B cell-driven dissemination.


Asunto(s)
Infecciones por Herpesviridae/virología , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/virología , Tropismo Viral , Replicación Viral , Animales , Linfocitos B/virología , Línea Celular , Células Endoteliales/virología , Genes Reporteros , Infecciones por Herpesviridae/patología , Estudios Longitudinales , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Rhadinovirus/genética , Rhadinovirus/crecimiento & desarrollo , Rhadinovirus/fisiología , Bazo/virología , Coloración y Etiquetado/métodos , Infecciones Tumorales por Virus/patología
16.
PLoS Pathog ; 8(2): e1002549, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22427750

RESUMEN

Gene expression of DNA viruses requires nuclear import of the viral genome. Human Adenoviruses (Ads), like most DNA viruses, encode factors within early transcription units promoting their own gene expression and counteracting cellular antiviral defense mechanisms. The cellular transcriptional repressor Daxx prevents viral gene expression through the assembly of repressive chromatin remodeling complexes targeting incoming viral genomes. However, it has remained unclear how initial transcriptional activation of the adenoviral genome is achieved. Here we show that Daxx mediated repression of the immediate early Ad E1A promoter is efficiently counteracted by the capsid protein VI. This requires a conserved PPxY motif in protein VI. Capsid proteins from other DNA viruses were also shown to activate the Ad E1A promoter independent of Ad gene expression and support virus replication. Our results show how Ad entry is connected to transcriptional activation of their genome in the nucleus. Our data further suggest a common principle for genome activation of DNA viruses by counteracting Daxx related repressive mechanisms through virion proteins.


Asunto(s)
Adenoviridae/genética , Proteínas de la Cápside/fisiología , Genoma Viral , Activación Transcripcional/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Secuencias de Aminoácidos/genética , Secuencias de Aminoácidos/fisiología , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Células Cultivadas , Proteínas Co-Represoras , Regulación Viral de la Expresión Génica , Genes Virales/fisiología , Aptitud Genética/fisiología , Genoma Viral/genética , Humanos , Chaperonas Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Transfección , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Virales/fisiología , Replicación Viral/genética
17.
PLoS Pathog ; 8(12): e1003062, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23271968

RESUMEN

The inhibition of death-receptor apoptosis is a conserved viral function. The murine cytomegalovirus (MCMV) gene M36 is a sequence and functional homologue of the human cytomegalovirus gene UL36, and it encodes an inhibitor of apoptosis that binds to caspase-8, blocks downstream signaling and thus contributes to viral fitness in macrophages and in vivo. Here we show a direct link between the inability of mutants lacking the M36 gene (ΔM36) to inhibit apoptosis, poor viral growth in macrophage cell cultures and viral in vivo fitness and virulence. ΔM36 grew poorly in RAG1 knockout mice and in RAG/IL-2-receptor common gamma chain double knockout mice (RAGγC(-/-)), but the depletion of macrophages in either mouse strain rescued the growth of ΔM36 to almost wild-type levels. This was consistent with the observation that activated macrophages were sufficient to impair ΔM36 growth in vitro. Namely, spiking fibroblast cell cultures with activated macrophages had a suppressive effect on ΔM36 growth, which could be reverted by z-VAD-fmk, a chemical apoptosis inhibitor. TNFα from activated macrophages synergized with IFNγ in target cells to inhibit ΔM36 growth. Hence, our data show that poor ΔM36 growth in macrophages does not reflect a defect in tropism, but rather a defect in the suppression of antiviral mediators secreted by macrophages. To the best of our knowledge, this shows for the first time an immune evasion mechanism that protects MCMV selectively from the antiviral activity of macrophages, and thus critically contributes to viral pathogenicity in the immunocompromised host devoid of the adaptive immune system.


Asunto(s)
Inmunidad Adaptativa , Inmunodeficiencia Variable Común/inmunología , Infecciones por Herpesviridae/inmunología , Macrófagos/inmunología , Muromegalovirus/inmunología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Línea Celular , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/patología , Inmunodeficiencia Variable Común/virología , Inhibidores de Cisteína Proteinasa/farmacología , Fibroblastos/inmunología , Fibroblastos/patología , Fibroblastos/virología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/patología , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Macrófagos/patología , Macrófagos/virología , Ratones , Ratones Noqueados , Muromegalovirus/genética , Proteínas Virales/genética , Proteínas Virales/inmunología
18.
PLoS Pathog ; 8(6): e1002728, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22685399

RESUMEN

There is increasing evidence for a connection between DNA replication and the expression of adjacent genes. Therefore, this study addressed the question of whether a herpesvirus origin of replication can be used to activate or increase the expression of adjacent genes. Cell lines carrying an episomal vector, in which reporter genes are linked to the murine cytomegalovirus (MCMV) origin of lytic replication (oriLyt), were constructed. Reporter gene expression was silenced by a histone-deacetylase-dependent mechanism, but was resolved upon lytic infection with MCMV. Replication of the episome was observed subsequent to infection, leading to the induction of gene expression by more than 1000-fold. oriLyt-based regulation thus provided a unique opportunity for virus-induced conditional gene expression without the need for an additional induction mechanism. This principle was exploited to show effective late trans-complementation of the toxic viral protein M50 and the glycoprotein gO of MCMV. Moreover, the application of this principle for intracellular immunization against herpesvirus infection was demonstrated. The results of the present study show that viral infection specifically activated the expression of a dominant-negative transgene, which inhibited viral growth. This conditional system was operative in explant cultures of transgenic mice, but not in vivo. Several applications are discussed.


Asunto(s)
Infecciones por Citomegalovirus/genética , Citomegalovirus/genética , Regulación Viral de la Expresión Génica/genética , Muromegalovirus/genética , Animales , Western Blotting , Genes Reporteros , Hibridación Fluorescente in Situ , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Células 3T3 NIH , Replicón/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
PLoS Pathog ; 8(9): e1002908, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22969428

RESUMEN

During viral infections cellular gene expression is subject to rapid alterations induced by both viral and antiviral mechanisms. In this study, we applied metabolic labeling of newly transcribed RNA with 4-thiouridine (4sU-tagging) to dissect the real-time kinetics of cellular and viral transcriptional activity during lytic murine cytomegalovirus (MCMV) infection. Microarray profiling on newly transcribed RNA obtained at different times during the first six hours of MCMV infection revealed discrete functional clusters of cellular genes regulated with distinct kinetics at surprising temporal resolution. Immediately upon virus entry, a cluster of NF-κB- and interferon-regulated genes was induced. Rapid viral counter-regulation of this coincided with a very transient DNA-damage response, followed by a delayed ER-stress response. Rapid counter-regulation of all three clusters indicated the involvement of novel viral regulators targeting these pathways. In addition, down-regulation of two clusters involved in cell-differentiation (rapid repression) and cell-cycle (delayed repression) was observed. Promoter analysis revealed all five clusters to be associated with distinct transcription factors, of which NF-κB and c-Myc were validated to precisely match the respective transcriptional changes observed in newly transcribed RNA. 4sU-tagging also allowed us to study the real-time kinetics of viral gene expression in the absence of any interfering virion-associated-RNA. Both qRT-PCR and next-generation sequencing demonstrated a sharp peak of viral gene expression during the first two hours of infection including transcription of immediate-early, early and even well characterized late genes. Interestingly, this was subject to rapid gene silencing by 5-6 hours post infection. Despite the rapid increase in viral DNA load during viral DNA replication, transcriptional activity of some viral genes remained remarkably constant until late-stage infection, or was subject to further continuous decline. In summary, this study pioneers real-time transcriptional analysis during a lytic herpesvirus infection and highlights numerous novel regulatory aspects of virus-host-cell interaction.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/genética , Interacciones Huésped-Patógeno/genética , Muromegalovirus/genética , Animales , Perfilación de la Expresión Génica/métodos , Genes Virales/genética , Infecciones por Herpesviridae/virología , Ratones , Análisis por Micromatrices , Familia de Multigenes/genética , Muromegalovirus/patogenicidad , Células 3T3 NIH , Regiones Promotoras Genéticas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/genética , Transcripción Genética/genética
20.
PLoS Pathog ; 8(2): e1002510, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22346748

RESUMEN

Cytomegaloviruses express large amounts of viral miRNAs during lytic infection, yet, they only modestly alter the cellular miRNA profile. The most prominent alteration upon lytic murine cytomegalovirus (MCMV) infection is the rapid degradation of the cellular miR-27a and miR-27b. Here, we report that this regulation is mediated by the ∼1.7 kb spliced and highly abundant MCMV m169 transcript. Specificity to miR-27a/b is mediated by a single, apparently optimized, miRNA binding site located in its 3'-UTR. This site is easily and efficiently retargeted to other cellular and viral miRNAs by target site replacement. Expression of the 3'-UTR of m169 by an adenoviral vector was sufficient to mediate its function, indicating that no other viral factors are essential in this process. Degradation of miR-27a/b was found to be accompanied by 3'-tailing and -trimming. Despite its dramatic effect on miRNA stability, we found this interaction to be mutual, indicating potential regulation of m169 by miR-27a/b. Most interestingly, three mutant viruses no longer able to target miR-27a/b, either due to miRNA target site disruption or target site replacement, showed significant attenuation in multiple organs as early as 4 days post infection, indicating that degradation of miR-27a/b is important for efficient MCMV replication in vivo.


Asunto(s)
Regiones no Traducidas 3'/genética , Infecciones por Citomegalovirus/virología , MicroARNs/metabolismo , Muromegalovirus/fisiología , ARN Viral/metabolismo , Replicación Viral/genética , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sitios de Unión , Línea Celular , Regulación hacia Abajo/genética , Regulación de la Expresión Génica/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , MicroARNs/genética , Muromegalovirus/genética , Mutación , Procesamiento Postranscripcional del ARN , Estabilidad del ARN/genética , ARN Viral/genética , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA