Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 98(6): e0000524, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38717113

RESUMEN

TRIM32 is often aberrantly expressed in many types of cancers. Kaposi's sarcoma-associated herpesvirus (KSHV) is linked with several human malignancies, including Kaposi's sarcoma and primary effusion lymphomas (PELs). Increasing evidence has demonstrated the crucial role of KSHV lytic replication in viral tumorigenesis. However, the role of TRIM32 in herpesvirus lytic replication remains unclear. Here, we reveal that the expression of TRIM32 is upregulated by KSHV in latency, and reactivation of KSHV lytic replication leads to the inhibition of TRIM32 in PEL cells. Strikingly, RTA, the master regulator of lytic replication, interacts with TRIM32 and dramatically promotes TRIM32 for degradation via the proteasome systems. Inhibition of TRIM32 induces cell apoptosis and in turn inhibits the proliferation and colony formation of KSHV-infected PEL cells and facilitates the reactivation of KSHV lytic replication and virion production. Thus, our data imply that the degradation of TRIM32 is vital for the lytic activation of KSHV and is a potential therapeutic target for KSHV-associated cancers. IMPORTANCE: TRIM32 is associated with many cancers and viral infections; however, the role of TRIM32 in viral oncogenesis remains largely unknown. In this study, we found that the expression of TRIM32 is elevated by Kaposi's sarcoma-associated herpesvirus (KSHV) in latency, and RTA (the master regulator of lytic replication) induces TRIM32 for proteasome degradation upon viral lytic reactivation. This finding provides a potential therapeutic target for KSHV-associated cancers.


Asunto(s)
Herpesvirus Humano 8 , Proteínas Inmediatas-Precoces , Proteolisis , Transactivadores , Factores de Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Activación Viral , Replicación Viral , Humanos , Apoptosis , Línea Celular , Herpesvirus Humano 8/crecimiento & desarrollo , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/patogenicidad , Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Inmediatas-Precoces/genética , Linfoma de Efusión Primaria/virología , Linfoma de Efusión Primaria/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Sarcoma de Kaposi/virología , Sarcoma de Kaposi/metabolismo , Transactivadores/metabolismo , Transactivadores/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Latencia del Virus
2.
J Virol ; 97(11): e0138923, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37888983

RESUMEN

IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is a cancer-causing human herpesvirus that establishes a persistent infection in humans. The lytic viral cycle plays a crucial part in lifelong infection as it is involved in the viral dissemination. The master regulator of the KSHV lytic replication cycle is the viral replication and transcription activator (RTA) protein, which is necessary and sufficient to push the virus from latency into the lytic phase. Thus, the identification of host factors utilized by RTA for controlling the lytic cycle can help to find novel targets that could be used for the development of antiviral therapies against KSHV. Using a proteomics approach, we have identified a novel interaction between RTA and the cellular E3 ubiquitin ligase complex RNF20/40, which we have shown to be necessary for promoting RTA-induced KSHV lytic cycle.


Asunto(s)
Herpesvirus Humano 8 , Interacciones Microbiota-Huesped , Proteínas Inmediatas-Precoces , Ubiquitina-Proteína Ligasas , Proteínas Virales , Activación Viral , Latencia del Virus , Replicación Viral , Humanos , Herpesvirus Humano 8/crecimiento & desarrollo , Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Unión Proteica , Proteómica , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo
3.
Nature ; 553(7689): 521-525, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29342139

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) causes Kaposi's sarcoma, a cancer that commonly affects patients with AIDS and which is endemic in sub-Saharan Africa. The KSHV capsid is highly pressurized by its double-stranded DNA genome, as are the capsids of the eight other human herpesviruses. Capsid assembly and genome packaging of herpesviruses are prone to interruption and can therefore be targeted for the structure-guided development of antiviral agents. However, herpesvirus capsids-comprising nearly 3,000 proteins and over 1,300 Å in diameter-present a formidable challenge to atomic structure determination and functional mapping of molecular interactions. Here we report a 4.2 Å resolution structure of the KSHV capsid, determined by electron-counting cryo-electron microscopy, and its atomic model, which contains 46 unique conformers of the major capsid protein (MCP), the smallest capsid protein (SCP) and the triplex proteins Tri1 and Tri2. Our structure and mutagenesis results reveal a groove in the upper domain of the MCP that contains hydrophobic residues that interact with the SCP, which in turn crosslinks with neighbouring MCPs in the same hexon to stabilize the capsid. Multiple levels of MCP-MCP interaction-including six sets of stacked hairpins lining the hexon channel, disulfide bonds across channel and buttress domains in neighbouring MCPs, and an interaction network forged by the N-lasso domain and secured by the dimerization domain-define a robust capsid that is resistant to the pressure exerted by the enclosed genome. The triplexes, each composed of two Tri2 molecules and a Tri1 molecule, anchor to the capsid floor via a Tri1 N-anchor to plug holes in the MCP network and rivet the capsid floor. These essential roles of the MCP N-lasso and Tri1 N-anchor are verified by serial-truncation mutageneses. Our proof-of-concept demonstration of the use of polypeptides that mimic the smallest capsid protein to inhibit KSHV lytic replication highlights the potential for exploiting the interaction hotspots revealed in our atomic structure to develop antiviral agents.


Asunto(s)
Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Microscopía por Crioelectrón , Herpesvirus Humano 8/crecimiento & desarrollo , Herpesvirus Humano 8/ultraestructura , Mutagénesis , Replicación Viral , Cápside/química , Cápside/metabolismo , Cápside/ultraestructura , Proteínas de la Cápside/química , Proteínas de la Cápside/ultraestructura , Disulfuros/metabolismo , Diseño de Fármacos , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestructura , Mutación , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Estabilidad Proteica , Replicación Viral/genética
4.
Semin Cell Dev Biol ; 111: 135-147, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32631785

RESUMEN

Oncogenic DNA viruses establish lifelong infections in humans, and they cause cancers, often in immunocompromised patients, despite anti-viral immune surveillance targeted against viral antigens. High-throughput sequencing techniques allowed the field to identify novel viral non-coding RNAs (ncRNAs). ncRNAs are ideal factors for DNA viruses to exploit; they are non-immunogenic to T cells, thus viral ncRNAs can manipulate host cells without evoking adaptive immune responses. Viral ncRNAs may still trigger the host innate immune response, but many viruses encode decoys/inhibitors to counter-act and evade recognition. In addition, ncRNAs can be secreted to the extracellular space and influence adjacent cells to create a pro-viral microenvironment. In this review, we present recent progress in understanding interactions between oncoviruses and ncRNAs including small and long ncRNAs, microRNAs, and recently identified viral circular RNAs. In addition, potential clinical applications for ncRNA will be discussed. Extracellular ncRNAs are suggested to be diagnostic and prognostic biomarkers and, with the realization of the importance of viral ncRNAs in tumorigenesis, approaches to target critical viral ncRNAs are emerging. Further understanding of viral utilization of ncRNAs will advance anti-viral therapeutics beyond conventional medication and vaccination.


Asunto(s)
Evasión Inmune/genética , MicroARNs/genética , Neoplasias/genética , ARN Circular/genética , ARN Largo no Codificante/genética , ARN Viral/genética , Virosis/genética , Alphapapillomavirus/genética , Alphapapillomavirus/crecimiento & desarrollo , Alphapapillomavirus/patogenicidad , Antivirales/uso terapéutico , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/patología , Regulación de la Expresión Génica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Herpesvirus Humano 8/patogenicidad , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/crecimiento & desarrollo , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Inmunidad Innata , MicroARNs/antagonistas & inhibidores , MicroARNs/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/virología , Oligonucleótidos Antisentido/uso terapéutico , ARN Circular/inmunología , ARN Largo no Codificante/inmunología , ARN Viral/inmunología , Transducción de Señal , Virosis/inmunología , Virosis/terapia , Virosis/virología
5.
J Virol ; 95(13): e0009621, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33853955

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic human gammaherpesvirus and the causative agent of Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease (MCD). During reactivation, viral genes are expressed in a temporal manner. These lytic genes encode transactivators, core replication proteins, or structural proteins. During reactivation, other viral factors that are required for lytic replication are expressed. The most abundant viral transcript is the long noncoding RNA (lncRNA) known as polyadenylated nuclear (PAN) RNA. lncRNAs have diverse functions, including the regulation of gene expression and the immune response. PAN possesses two main cis-acting elements, the Mta response element (MRE) and the expression and nuclear retention element (ENE). While PAN has been demonstrated to be required for efficient viral replication, the function of these elements within PAN remains unclear. Our goal was to determine if the ENE of PAN is required in the context of infection. A KSHV bacmid containing a deletion of the 79-nucleotide (nt) ENE in PAN was generated to assess the effects of the ENE during viral replication. Our studies demonstrated that the ENE is not required for viral DNA synthesis, lytic gene expression, or the production of infectious virus. Although the ENE is not required for viral replication, we found that the ENE functions to retain PAN in the nucleus, and the absence of the ENE results in an increased accumulation of PAN in the cytoplasm. Furthermore, open reading frame 59 (ORF59), LANA, ORF57, H1.4, and H2A still retain the ability to bind to PAN in the absence of the ENE. Together, our data highlight how the ENE affects the nuclear retention of PAN but ultimately does not play an essential role during lytic replication. Our data suggest that PAN may have other functional domains apart from the ENE. IMPORTANCE KSHV is an oncogenic herpesvirus that establishes latency and exhibits episodes of reactivation. KSHV disease pathologies are most often associated with the lytic replication of the virus. PAN RNA is the most abundant viral transcript during the reactivation of KSHV and is required for viral replication. Deletion and knockdown of PAN resulted in defects in viral replication and reduced virion production in the absence of PAN RNA. To better understand how the cis elements within PAN may contribute to its function, we investigated if the ENE of PAN was necessary for viral replication. Although the ENE had previously been extensively studied with both biochemical and in vitro approaches, this is the first study to demonstrate the role of the ENE in the context of infection and that the ENE of PAN is not required for the lytic replication of KSHV.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Herpesvirus Humano 8/genética , ARN Largo no Codificante/genética , Activación Viral/genética , Latencia del Virus/genética , Enfermedad de Castleman/virología , Línea Celular Tumoral , Células HEK293 , Herpesvirus Humano 8/fisiología , Humanos , ARN Mensajero/genética , ARN Nuclear/genética , Sarcoma de Kaposi/virología , Replicación Viral/genética
6.
Adv Exp Med Biol ; 1225: 127-135, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32030652

RESUMEN

Epstein Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV) constitute the human γ-herpesviruses and two of the seven human tumor viruses. In addition to their viral oncogenes that primarily belong to the latent infection programs of these viruses, they encode proteins that condition the microenvironment. Many of these are early lytic gene products and are only expressed in a subset of infected cells of the tumor mass. In this chapter I will describe their function and the evidence that targeting them in addition to the latent oncogenes could be beneficial for the treatment of EBV- and KSHV-associated malignancies.


Asunto(s)
Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 8/crecimiento & desarrollo , Herpesvirus Humano 8/patogenicidad , Neoplasias/tratamiento farmacológico , Neoplasias/virología , Oncogenes , Microambiente Tumoral , Replicación Viral , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/efectos de los fármacos , Herpesvirus Humano 8/genética , Humanos , Oncogenes/efectos de los fármacos , Replicación Viral/efectos de los fármacos
7.
J Virol ; 92(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30158293

RESUMEN

Minichromosome maintenance proteins (MCMs) play an important role in DNA replication by binding to the origins as helicase and recruiting polymerases for DNA synthesis. During the S phase, MCM complex is loaded to limit DNA replication once per cell cycle. We identified MCMs as ORF59 binding partners in our protein pulldown assays, which led us to hypothesize that this interaction influences DNA replication. ORF59's interactions with MCMs were confirmed in both endogenous and overexpression systems, which showed its association with MCM3, MCM4, MCM5, and MCM6. Interestingly, MCM6 interacted with both the N- and C-terminal domains of ORF59, and its depletion in BCBL-1 and BC3 cells led to an increase in viral genome copies, viral late gene transcripts, and virion production compared to the control cells following reactivation. MCMs perform their function by loading onto the replication competent DNA, and one means of regulating chromatin loading/unloading, in addition to enzymatic activity of the MCM complex, is by posttranslational modifications, including phosphorylation of these factors. Interestingly, a hypophosphorylated form of MCM3, which is associated with reduced loading onto the chromatin, was detected during lytic reactivation and correlated with its inability to associate with histones in reactivated cells. Additionally, chromatin immunoprecipitation showed lower levels of MCM3 and MCM4 association at cellular origins of replication and decreased levels of cellular DNA synthesis in cells undergoing reactivation. Taken together, these findings suggest a mechanism in which KSHV ORF59 disrupts the assembly and functions of MCM complex to stall cellular DNA replication and promote viral replication.IMPORTANCE KSHV is the causative agent of various lethal malignancies affecting immunocompromised individuals. Both lytic and latent phases of the viral life cycle contribute to the progression of these cancers. A better understanding of how viral proteins disrupt functions of a normal healthy cell to cause oncogenesis is warranted. One crucial lytic protein produced early during lytic reactivation is the multifunctional ORF59. In this report, we elucidated an important role of ORF59 in manipulating the cellular environment conducive for viral DNA replication by deregulating the normal functions of the host MCM proteins. ORF59 binds to specific MCMs and sequesters them away from replication origins in order to sabotage cellular DNA replication. Blocking cellular DNA replication ensures that cellular resources are utilized for transcription and replication of viral DNA.


Asunto(s)
División Celular/genética , Replicación del ADN/genética , Herpesvirus Humano 8/genética , Proteínas de Mantenimiento de Minicromosoma/genética , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Sarcoma de Kaposi/genética , Proteínas Virales/genética , Acetiltransferasas/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Células HEK293 , Herpesvirus Humano 8/crecimiento & desarrollo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Componente 4 del Complejo de Mantenimiento de Minicromosoma/genética , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/genética , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/virología , Activación Viral/genética
8.
PLoS Pathog ; 13(1): e1006143, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28046107

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV) causes several tumors and hyperproliferative disorders. Hypoxia and hypoxia-inducible factors (HIFs) activate latent and lytic KSHV genes, and several KSHV proteins increase the cellular levels of HIF. Here, we used RNA sequencing, qRT-PCR, Taqman assays, and pathway analysis to explore the miRNA and mRNA response of uninfected and KSHV-infected cells to hypoxia, to compare this with the genetic changes seen in chronic latent KSHV infection, and to explore the degree to which hypoxia and KSHV infection interact in modulating mRNA and miRNA expression. We found that the gene expression signatures for KSHV infection and hypoxia have a 34% overlap. Moreover, there were considerable similarities between the genes up-regulated by hypoxia in uninfected (SLK) and in KSHV-infected (SLKK) cells. hsa-miR-210, a HIF-target known to have pro-angiogenic and anti-apoptotic properties, was significantly up-regulated by both KSHV infection and hypoxia using Taqman assays. Interestingly, expression of KSHV-encoded miRNAs was not affected by hypoxia. These results demonstrate that KSHV harnesses a part of the hypoxic cellular response and that a substantial portion of hypoxia-induced changes in cellular gene expression are induced by KSHV infection. Therefore, targeting hypoxic pathways may be a useful way to develop therapeutic strategies for KSHV-related diseases.


Asunto(s)
Hipoxia de la Célula/genética , Regulación de la Expresión Génica/genética , Herpesvirus Humano 8/crecimiento & desarrollo , MicroARNs/genética , Sarcoma de Kaposi/genética , Secuencia de Bases , Línea Celular Tumoral , Biología Computacional , Células Endoteliales/patología , Células Endoteliales/virología , Herpesvirus Humano 8/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , MicroARNs/biosíntesis , Sarcoma de Kaposi/virología , Análisis de Secuencia de ARN
10.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 1-11, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27751885

RESUMEN

Members of the herpesviral family use multiple strategies to hijack infected host cells and exploit cellular signaling for their pathogenesis and latent infection. Among the most intriguing weapons in the arsenal of pathogenic herpesviruses are the constitutively active virally-encoded G protein-coupled receptors (vGPCRs). Even though vGPCRs contribute to viral pathogenesis such as immune evasion and proliferative disorders, the molecular details of how vGPCRs continuously activate cellular signaling are largely unknown. Here, we report that the vGPCR of Herpesvirus saimiri (HVS), an oncogenic γ2-herpesvirus, constitutively activates T cells via a heteromeric interaction with cellular CXCR4. Constitutive T cell activation also occurs with expression of the vGPCR of Kaposi's sarcoma-associated herpesvirus (KSHV), but not the vGPCR of Epstein-Barr virus. Expression of HVS vGPCR down-regulated the surface expression of CXCR4 but did not induce the degradation of the chemokine receptor, suggesting that vGPCR/CXCR4 signaling continues in cytosolic compartments. The physical association of vGPCR with CXCR4 was demonstrated by proximity ligation assay as well as immunoprecipitation. Interestingly, the constitutive activation of T cells by HVS vGPCR is independent of proximal T cell receptor (TCR) signaling molecules, such as TCRß, Lck, and ZAP70, whereas CXCR4 silencing by shRNA abolished T cell activation by vGPCRs of HVS and KSHV. Furthermore, previously identified inactive vGPCR mutants failed to interact with CXCR4. These findings on the positive cooperativity of vGPCR with cellular CXCR4 in T cell activation extend our current understanding of the molecular mechanisms of vGPCR function and highlight the importance of heteromerization for GPCR activity.


Asunto(s)
Herpesvirus Saimiriino 2/metabolismo , Herpesvirus Humano 8/metabolismo , Receptores CXCR4/genética , Receptores de Quimiocina/genética , Linfocitos T/virología , Regulación de la Expresión Génica , Células HEK293 , Herpesvirus Saimiriino 2/genética , Herpesvirus Saimiriino 2/crecimiento & desarrollo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Humanos , Activación de Linfocitos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/inmunología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Cultivo Primario de Células , Unión Proteica , Multimerización de Proteína , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteína Tirosina Quinasa ZAP-70/genética , Proteína Tirosina Quinasa ZAP-70/inmunología , Proteína Tirosina Quinasa ZAP-70/metabolismo
11.
J Virol ; 91(19)2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28747501

RESUMEN

Latency-associated nuclear antigen (LANA) is a multifunctional protein encoded by members of the Rhadinovirus genus of gammaherpesviruses. Studies using murine gammaherpesvirus 68 (MHV68) demonstrated that LANA is important for acute replication, latency establishment, and reactivation in vivo Despite structural similarities in their DNA-binding domains (DBDs), LANA homologs from Kaposi sarcoma-associated herpesvirus (KSHV) and MHV68 exhibit considerable sequence divergence. We sought to determine if KSHV and MHV68 LANA homologs are functionally interchangeable. We generated an MHV68 virus that encodes KSHV LANA (kLANA) in place of MHV68 LANA (mLANA) and evaluated the virus's capacity to replicate, establish and maintain latency, and reactivate. kLANA knock-in (KLKI) MHV68 was replication competent in vitro and in vivo but exhibited slower growth kinetics and lower titers than wild-type (WT) MHV68. Following inoculation of mice, KLKI MHV68 established and maintained latency in splenocytes and peritoneal cells but did not reactivate efficiently ex vivo kLANA repressed the MHV68 promoter for ORF50, the gene that encodes the major lytic transactivator protein RTA, while mLANA did not, suggesting a likely mechanism for the KLKI MHV68 phenotypes. Bypassing this repression by providing MHV68 RTA in trans rescued KLKI MHV68 replication in tissue culture and enabled detection of KLKI MHV68 reactivation ex vivo These data demonstrate that kLANA and mLANA are functionally interchangeable for establishment and maintenance of latency and suggest that repression of lytic replication by kLANA, as previously shown with KSHV, is a kLANA-specific function that is transferable to MHV68.IMPORTANCE Kaposi sarcoma-associated herpesvirus (KSHV) and murine gammaherpesvirus 68 (MHV68) are members of the Rhadinovirus genus of gammaherpesviruses. These viruses establish lifelong infections that place their respective human and murine hosts at risk for cancer. Latency-associated nuclear antigen (LANA) is a conserved Rhadinovirus protein that is necessary for long-term chronic infection by these viruses. To better understand the conserved functions performed by LANA homologs, we generated a recombinant MHV68 virus that encodes the KSHV LANA protein in place of the MHV68 LANA homolog. We determined that the KSHV LANA protein is capable of supporting MHV68 latency in a mouse model of chronic infection but also functions to repress viral replication. This work describes an in vivo model system for defining evolutionarily conserved and divergent functions of LANA homologs in Rhadinovirus infection and disease.


Asunto(s)
Antígenos Virales/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Proteínas Inmediatas-Precoces/genética , Proteínas Nucleares/genética , Rhadinovirus/crecimiento & desarrollo , Transactivadores/genética , Latencia del Virus/genética , Células 3T3 , Animales , Antígenos Virales/biosíntesis , Línea Celular , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/biosíntesis , Regiones Promotoras Genéticas/genética , Rhadinovirus/genética , Rhadinovirus/metabolismo
12.
Apoptosis ; 22(9): 1098-1115, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28653223

RESUMEN

Kaposi's sarcoma associated herpes virus (KSHV) infected primary effusion lymphoma (PEL) is a rare aggressive form of non-Hodgkin's lymphoma of B cells. KSHV latent and lytic antigens modulate several host cellular signalling pathways especially mammalian target of rapamycin (mTOR), STAT-3 and nuclear factor-kappa B (NF-κB) for rapid tumor progression and immune evasion. Current chemotherapeutic strategies are becoming ineffective as they kill only dividing cells and inefficient to target molecular pathways crucial for active virus replication and its survival. In this study, we evaluated the efficacy of everolimus, an mTOR inhibitor in inducing apoptosis of PEL cells. Dose-dependent treatment of everolimus triggered mitochondria-mediated caspase-dependent apoptosis in PEL cells. Everolimus downregulated KSHV latent antigen expression with concurrent blocking of lytic reactivation for active virus replication. Everolimus also inhibited latent antigen mediated constitutively active STAT-3 and NF-κB signalling. We co-cultured everolimus treated PEL cells with immature dendritic cells and found activation of dendritic cells with increase in surface expression of CD86 and HLA-DR. As everolimus targets and disrupts KSHV antigens as well as antigen facilitated multiple signalling pathways necessary for KSHV survival and maintenance of infection with synchronised boosting of immune system against viral infection, it can be a better therapeutic approach towards treatment of PEL.


Asunto(s)
Apoptosis/efectos de los fármacos , Everolimus/farmacología , Herpesvirus Humano 8/efectos de los fármacos , Linfoma de Efusión Primaria/virología , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Relación Dosis-Respuesta a Droga , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Herpesvirus Humano 8/crecimiento & desarrollo , Humanos , Linfoma de Efusión Primaria/tratamiento farmacológico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
13.
J Virol ; 89(3): 1688-702, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25410858

RESUMEN

UNLABELLED: The Kaposi's sarcoma-associated herpesvirus (KSHV) ORF57 gene product is essential for lytic KSHV replication and virion production. Recombinant ORF57-null mutants fail to accumulate several lytic cycle mRNAs at wild-type levels, leading to decreased production of lytic proteins necessary for efficient replication. Several mechanisms by which ORF57 may enhance expression of lytic KSHV mRNAs have been proposed, including mRNA stabilization, mRNA nuclear export, increased polyadenylation, and transcriptional activation. ORF57 activity is also gene specific, with some genes being highly dependent on ORF57, whereas others are relatively independent. Most experiments have utilized transfection models for ORF57 and have not systematically examined the gene specificity and potential mechanisms of action of ORF57 in the context of KSHV-infected cells. In this study, the KSHV genes that are most highly upregulated by ORF57 during KSHV lytic replication were identified by a combination of high-throughput deep RNA sequencing, quantitative PCR, Northern blotting, and rapid amplification of cDNA ends methods. Comparison of gene expression from a ΔORF57 KSHV recombinant, a rescued ΔORF57 KSHV recombinant, and wild-type KSHV revealed that two clusters of lytic genes are most highly dependent on ORF57 for efficient expression. Despite contiguous location in the genome and shared polyadenylation of several of the ORF57-dependent genes, ORF57 regulation was promoter and polyadenylation signal independent, suggesting that the mRNAs are stabilized by ORF57. The eight genes identified to critically require ORF57 belong to both early and late lytic temporal classes, and seven are involved in DNA replication, virion assembly, or viral infectivity, explaining the essential role of ORF57 in infectious KSHV production. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is a human herpesvirus involved in the causation of several human cancers. The KSHV ORF57 protein is required for KSHV to replicate and produce infectious virus. We have identified several KSHV genes whose expression is highly dependent on ORF57 and shown that ORF57 increases expression of these genes specifically. These genes code for proteins that are required for the virus to replicate its DNA and to infect other cells. Identifying the targets and mechanism of action of ORF57 provides further approaches to discover antiviral therapy.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/fisiología , Proteínas Virales/metabolismo , Ensamble de Virus , Internalización del Virus , Liberación del Virus , Replicación Viral , Línea Celular , Eliminación de Gen , Perfilación de la Expresión Génica , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Humanos , Estabilidad del ARN , Proteínas Virales/genética
14.
J Virol ; 89(12): 6164-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25810547

RESUMEN

Ribosome profiling is an emerging technique that uses deep sequencing to monitor translation in live cells. Studies using ribosome profiling have already provided novel insights into the identities and amounts of the proteins being produced in cells, as well as novel insights into the mechanism of protein synthesis and translation regulation. Application of ribosome profiling to cells infected with human cytomegalovirus and Kaposi's sarcoma-associated herpesvirus revealed unanticipated complexity in the coding capacity of herpesviruses. Here, I discuss these results and how the application of ribosome profiling to cells infected with other viruses can reveal novel insights into the process of infection.


Asunto(s)
Citomegalovirus/crecimiento & desarrollo , Perfilación de la Expresión Génica/métodos , Herpesvirus Humano 8/crecimiento & desarrollo , Biología Molecular/métodos , Biosíntesis de Proteínas , Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Humanos , ARN Mensajero/genética , Ribosomas/genética
15.
J Virol ; 89(14): 7097-107, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25926644

RESUMEN

UNLABELLED: Latent Kaposi's sarcoma-associated herpesvirus (KSHV) genomes encode a homolog of cellular FLICE-inhibitory proteins (termed v-FLIP) that activates NF-κB and can trigger important proinflammatory and antiapoptotic changes in latently infected cells. The protein is present at very low levels in infection and has generally been difficult to efficiently express in recombinant vectors. Here we show that codon usage in the v-FLIP gene is strikingly suboptimal. Optimization of codon use in expression vectors, as expected, restores efficient protein expression. Surprisingly, however, it also dramatically increases the steady-state level of v-FLIP mRNA, at least in part by increasing mRNA stability. When codon-optimized v-FLIP sequences are reintroduced into intact KSHV genomes, the resulting virus expresses readily detectable monocistronic v-FLIP mRNAs that are undetectable in wild-type (WT) infection by blot hybridization, suggesting that such RNAs are in fact transcribed in WT infection but fail to accumulate. The overexpression of v-FLIP by codon-optimized latent genomes results in a 5- to 7-fold decrement in virus production following lytic induction, indicating that maximizing NF-κB signaling is deleterious to induction. These studies provide a clear explanation for the evolution of inefficient codon usage in this gene and point to a strong connection between translational efficiency and RNA accumulation in mammalian cells. IMPORTANCE: This study reports that inefficient codon usage in a herpesviral gene is strikingly correlated with the inability of its mRNA to accumulate in cells; correction of efficient translatability restores RNA abundance. A similar correlation has been reported in yeast species, but the mechanisms operating in mammalian cells appear substantially different.


Asunto(s)
Codón , Expresión Génica , Herpesvirus Humano 8/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Línea Celular , Herpesvirus Humano 8/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Humanos , FN-kappa B/metabolismo , Estabilidad del ARN
16.
J Virol ; 88(14): 8028-44, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24807711

RESUMEN

The lytic cycles of Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) are induced in cell culture by sodium butyrate (NaB), a short-chain fatty acid (SCFA) histone deacetylase (HDAC) inhibitor. Valproic acid (VPA), another SCFA and an HDAC inhibitor, induces the lytic cycle of KSHV but blocks EBV lytic reactivation. To explore the hypothesis that structural differences between NaB and VPA account for their functional effects on the two related viruses, we investigated the capacity of 16 structurally related short- and medium-chain fatty acids to promote or prevent lytic cycle reactivation. SCFAs differentially affected EBV and KSHV reactivation. KSHV was reactivated by all SCFAs that are HDAC inhibitors, including phenylbutyrate. However, several fatty acid HDAC inhibitors, such as isobutyrate and phenylbutyrate, did not reactivate EBV. Reactivation of KSHV lytic transcripts could not be blocked completely by any fatty acid tested. In contrast, several medium-chain fatty acids inhibited lytic activation of EBV. Fatty acids that blocked EBV reactivation were more lipophilic than those that activated EBV. VPA blocked activation of the BZLF1 promoter by NaB but did not block the transcriptional function of ZEBRA. VPA also blocked activation of the DNA damage response that accompanies EBV lytic cycle activation. Properties of SCFAs in addition to their effects on chromatin are likely to explain activation or repression of EBV. We concluded that fatty acids stimulate the two related human gammaherpesviruses to enter the lytic cycle through different pathways. Importance: Lytic reactivation of EBV and KSHV is needed for persistence of these viruses and plays a role in carcinogenesis. Our direct comparison highlights the mechanistic differences in lytic reactivation between related human oncogenic gammaherpesviruses. Our findings have therapeutic implications, as fatty acids are found in the diet and produced by the human microbiota. Small-molecule inducers of the lytic cycle are desired for oncolytic therapy. Inhibition of viral reactivation, alternatively, may prove useful in cancer treatment. Overall, our findings contribute to the understanding of pathways that control the latent-to-lytic switch and identify naturally occurring molecules that may regulate this process.


Asunto(s)
Ácidos Grasos/metabolismo , Herpesvirus Humano 4/fisiología , Herpesvirus Humano 8/fisiología , Activación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/virología , Línea Celular , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 8/crecimiento & desarrollo , Inhibidores de Histona Desacetilasas/metabolismo , Humanos
17.
BMC Microbiol ; 15: 74, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25887745

RESUMEN

BACKGROUND: Human Herpesvirus 8 (HHV8), the causative agent of Kaposi's sarcoma, induces an intense modification of lipid metabolism and enhances the angiogenic process in endothelial cells. In the present study, neutral lipid (NL) metabolism and angiogenesis were investigated in HHV8-infected HUVEC cells. The viral replication phases were verified by rtPCR and also by K8.1 and LANA immunostaining. RESULTS: Lipid droplets (Nile Red) were higher in all phases and NL staining (LipidTOX) combined with viral-antigen detection (immunofluorescence) demonstrated a NL content increase in infected cells. In particular, triglyceride synthesis increases in the lytic phase, whereas cholesteryl ester synthesis rises in the latent one. Moreover, the inhibition of cholesterol esterification reduces neo-tubule formation mainly in latently infected cells. CONCLUSIONS: We suggest that a reprogramming of cholesteryl ester metabolism is involved in regulating neo-angiogenesis in HHV8-infected cells and plays a likely role in the high metastatic potential of derived-tumours.


Asunto(s)
Herpesvirus Humano 8/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Células Endoteliales de la Vena Umbilical Humana/química , Células Endoteliales de la Vena Umbilical Humana/virología , Lípidos/análisis , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Neovascularización Patológica/virología
18.
PLoS Pathog ; 8(9): e1002927, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028325

RESUMEN

Kaposi's Sarcoma (KS), caused by Kaposi's Sarcoma Herpesvirus (KSHV), is a highly vascularised angiogenic tumor of endothelial cells, characterized by latently KSHV-infected spindle cells and a pronounced inflammatory infiltrate. Several KSHV proteins, including LANA-1 (ORF73), vCyclin (ORF72), vGPCR (ORF74), vIL6 (ORF-K2), vCCL-1 (ORF-K6), vCCL-2 (ORF-K4) and K1 have been shown to exert effects that can lead to the proliferation and atypical differentiation of endothelial cells and/or the secretion of cytokines with angiogenic and inflammatory properties (VEGF, bFGF, IL6, IL8, GROα, and TNFß). To investigate a role of the KSHV K15 protein in KSHV-mediated angiogenesis, we carried out a genome wide gene expression analysis on primary endothelial cells infected with KSHV wildtype (KSHVwt) and a KSHV K15 deletion mutant (KSHVΔK15). We found RCAN1/DSCR1 (Regulator of Calcineurin 1/Down Syndrome critical region 1), a cellular gene involved in angiogenesis, to be differentially expressed in KSHVwt- vs KSHVΔK15-infected cells. During physiological angiogenesis, expression of RCAN1 in endothelial cells is regulated by VEGF (vascular endothelial growth factor) through a pathway involving the activation of PLCγ1, Calcineurin and NFAT1. We found that K15 directly recruits PLCγ1, and thereby activates Calcineurin/NFAT1-dependent RCAN1 expression which results in the formation of angiogenic tubes. Primary endothelial cells infected with KSHVwt form angiogenic tubes upon activation of the lytic replication cycle. This effect is abrogated when K15 is deleted (KSHVΔK15) or silenced by an siRNA targeting the K15 expression. Our study establishes K15 as one of the KSHV proteins that contribute to KSHV-induced angiogenesis.


Asunto(s)
Herpesvirus Humano 8/metabolismo , Células Endoteliales de la Vena Umbilical Humana/virología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Musculares/metabolismo , Neovascularización Patológica/virología , Fosfolipasa C gamma/metabolismo , Proteínas Virales/metabolismo , Inductores de la Angiogénesis , Animales , Calcineurina/metabolismo , Línea Celular , Chlorocebus aethiops , Proteínas de Unión al ADN , Células HEK293 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Datos de Secuencia Molecular , Proteínas Musculares/genética , Factores de Transcripción NFATC/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Sarcoma de Kaposi/virología , Eliminación de Secuencia , Células Vero , Proteínas Virales/genética
19.
PLoS Pathog ; 8(1): e1002479, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22253595

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is tightly linked to at least two lymphoproliferative disorders, primary effusion lymphoma (PEL) and multicentric Castleman's disease (MCD). However, the development of KSHV-mediated lymphoproliferative disease is not fully understood. Here, we generated two recombinant KSHV viruses deleted for the first RBP-Jκ binding site (RTA(1st)) and all three RBP-Jκ binding sites (RTA(all)) within the RTA promoter. Our results showed that RTA(1st) and RTA(all) recombinant viruses possess increased viral latency and a decreased capability for lytic replication in HEK 293 cells, enhancing colony formation and proliferation of infected cells. Furthermore, recombinant RTA(1st) and RTA(all) viruses showed greater infectivity in human peripheral blood mononuclear cells (PBMCs) relative to wt KSHV. Interestingly, KSHV BAC36 wt, RTA(1st) and RTA(all) recombinant viruses infected both T and B cells and all three viruses efficiently infected T and B cells in a time-dependent manner early after infection. Also, the capability of both RTA(1st) and RTA(all) recombinant viruses to infect CD19+ B cells was significantly enhanced. Surprisingly, RTA(1st) and RTA(all) recombinant viruses showed greater infectivity for CD3+ T cells up to 7 days. Furthermore, studies in Telomerase-immortalized human umbilical vein endothelial (TIVE) cells infected with KSHV corroborated our data that RTA(1st) and RTA(all) recombinant viruses have enhanced ability to persist in latently infected cells with increased proliferation. These recombinant viruses now provide a model to explore early stages of primary infection in human PBMCs and development of KSHV-associated lymphoproliferative diseases.


Asunto(s)
Proliferación Celular , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Regiones Promotoras Genéticas , Transactivadores/genética , Latencia del Virus/genética , Sitios de Unión/genética , Células Cultivadas , Regulación Viral de la Expresión Génica , Células HEK293 , Infecciones por Herpesviridae/complicaciones , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/crecimiento & desarrollo , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/química , Trastornos Linfoproliferativos/etiología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/virología , Regiones Promotoras Genéticas/genética , Unión Proteica , Transactivadores/metabolismo
20.
Arch Virol ; 159(9): 2377-86, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24777829

RESUMEN

Kaposi's sarcoma (KS) is a vascular tumor, and KS spindle cells express endothelial-cell-specific markers. Generally, it is believed that KS originates from endothelial cells. However, as various mesodermal-derived tissue markers are also expressed in KS spindle cells, the exact origin of KS still needs to be elucidated. Here, Kaposi's sarcoma-associated herpesvirus (KSHV) was used to infect human mesenchymal stem cells derived from bone marrow (hMSC-bm), and we investigated the angiogenic properties of these cells, which are one of the most important pathologic features of KS. KSHV-infected hMSC-bm showed latent infection and increased tube formation activity in vitro. In addition, the expression of endothelial-cell-specific markers and a growth factor that affects the angiogenesis of endothelial cells was induced in KSHV-infected cells. This study suggests that human mesenchymal stem cells might have important roles in KS pathogenesis.


Asunto(s)
Herpesvirus Humano 8/crecimiento & desarrollo , Células Madre Mesenquimatosas/virología , Neovascularización Fisiológica , Biomarcadores/análisis , Células Cultivadas , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA