Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 649
Filtrar
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.
Cell Host Microbe ; 32(5): 755-767.e4, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38653242

RESUMEN

Kaposi sarcoma (KS) is the most common cancer in persons living with HIV. It is caused by KS-associated herpesvirus (KSHV). There exists no animal model for KS. Pronuclear injection of the 170,000-bp viral genome induces early-onset, aggressive angiosarcoma in transgenic mice. The tumors are histopathologically indistinguishable from human KS. As in human KS, all tumor cells express the viral latency-associated nuclear antigen (LANA). The tumors transcribe most viral genes, whereas endothelial cells in other organs only transcribe the viral latent genes. The tumor cells are of endothelial lineage and exhibit the same molecular pattern of pathway activation as KS, namely phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR, interleukin-10 (IL-10), and vascular endothelial growth factor (VEGF). The KSHV-induced tumors are more aggressive than Ha-ras-induced angiosarcomas. Overall survival is increased by prophylactic ganciclovir. Thus, whole-virus KSHV-transgenic mice represent an accurate model for KS and open the door for the genetic dissection of KS pathogenesis and evaluation of therapies, including vaccines.


Asunto(s)
Modelos Animales de Enfermedad , Hemangiosarcoma , Herpesvirus Humano 8 , Ratones Transgénicos , Sarcoma de Kaposi , Animales , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidad , Ratones , Hemangiosarcoma/virología , Hemangiosarcoma/genética , Hemangiosarcoma/patología , Sarcoma de Kaposi/virología , Sarcoma de Kaposi/patología , Genoma Viral , Humanos , Antígenos Virales/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Ganciclovir/uso terapéutico , Ganciclovir/farmacología , Interleucina-10/genética
3.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35177478

RESUMEN

The role of N6-methyladenosine (m6A) modifications has increasingly been associated with a diverse set of roles in modulating viruses and influencing the outcomes of viral infection. Here, we report that the landscape of m6A deposition is drastically shifted during Kaposi's sarcoma-associated herpesvirus (KSHV) lytic infection for both viral and host transcripts. In line with previous reports, we also saw an overall decrease in host methylation in favor of viral messenger RNA (mRNA), along with 5' hypomethylation and 3' hypermethylation. During KSHV lytic infection, a major shift in overall mRNA abundance is driven by the viral endoribonuclease SOX, which induces the decay of greater than 70% of transcripts. Here, we reveal that interlukin-6 (IL-6) mRNA, a well-characterized, SOX-resistant transcript, is m6A modified during lytic infection. Furthermore, we show that this modification falls within the IL-6 SOX resistance element, an RNA element in the IL-6 3' untranslated region (UTR) that was previously shown to be sufficient for protection from SOX cleavage. We show that the presence of this m6A modification is essential to confer SOX resistance to the IL-6 mRNA. We next show that this modification recruits the m6A reader YTHDC2 and found that YTHDC2 is necessary for the escape of the IL-6 transcript. These results shed light on how the host cell has evolved to use RNA modifications to circumvent viral manipulation of RNA fate during KSHV infection.


Asunto(s)
Endorribonucleasas/metabolismo , ARN Helicasas/metabolismo , Estabilidad del ARN/fisiología , Adenosina/análogos & derivados , Adenosina/genética , Adenosina/metabolismo , Línea Celular Tumoral , Endorribonucleasas/genética , Expresión Génica/genética , Regulación Viral de la Expresión Génica/genética , Células HEK293 , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Metilación , ARN Helicasas/genética , Estabilidad del ARN/genética , ARN Mensajero/metabolismo , ARN Viral/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
4.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34768838

RESUMEN

Epstein-Barr Virus (EBV) and Kaposi's sarcoma associated-herpesvirus (KSHV) are γ-herpesviruses that belong to the Herpesviridae family. EBV infections are linked to the onset and progression of several diseases, such as Burkitt lymphoma (BL), nasopharyngeal carcinoma (NPC), and lymphoproliferative malignancies arising in post-transplanted patients (PTDLs). KSHV, an etiologic agent of Kaposi's sarcoma (KS), displays primary effusion lymphoma (PEL) and multicentric Castleman disease (MCD). Many therapeutics, such as bortezomib, CHOP cocktail medications, and natural compounds (e.g., quercetin or curcumin), are administrated to patients affected by γ-herpesvirus infections. These drugs induce apoptosis and autophagy, inhibiting the proliferative and cell cycle progression in these malignancies. In the last decade, many studies conducted by scientists and clinicians have indicated that nanotechnology and nanomedicine could improve the outcome of several treatments in γ-herpesvirus-associated diseases. Some drugs are entrapped in nanoparticles (NPs) expressed on the surface area of polyethylene glycol (PEG). These NPs move to specific tissues and exert their properties, releasing therapeutics in the cell target. To treat EBV- and KSHV-associated diseases, many studies have been performed in vivo and in vitro using virus-like particles (VPLs) engineered to maximize antigen and epitope presentations during immune response. NPs are designed to improve therapeutic delivery, avoiding dissolving the drugs in toxic solvents. They reduce the dose-limiting toxicity and reach specific tissue areas. Several attempts are ongoing to synthesize and produce EBV vaccines using nanosystems.


Asunto(s)
Gammaherpesvirinae/metabolismo , Infecciones por Herpesviridae/terapia , Nanotecnología/tendencias , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/terapia , Gammaherpesvirinae/genética , Gammaherpesvirinae/patogenicidad , Herpesviridae/metabolismo , Herpesviridae/patogenicidad , Infecciones por Herpesviridae/patología , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/patogenicidad , Humanos , Nanopartículas/uso terapéutico , Nanotecnología/métodos , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/terapia , Proteínas Virales/metabolismo , Replicación Viral/fisiología
5.
Nucleic Acids Res ; 49(16): 9389-9403, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34387695

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is a key DNA sensor that detects aberrant cytosolic DNA arising from pathogen invasions or genotoxic stresses. Upon binding to DNA, cGAS is activated and catalyzes the synthesis of cyclic GMP-AMP (cGAMP), which induces potent antimicrobial and antitumor responses. Kaposi sarcoma-associated herpesvirus (KSHV) is a human DNA tumor virus that causes Kaposi sarcoma and several other malignancies. We previously reported that KSHV inhibitor of cGAS (KicGAS) encoded by ORF52, inhibits cGAS enzymatic activity, but the underlying mechanisms remained unclear. To define the inhibitory mechanisms, here we performed in-depth biochemical and functional characterizations of KicGAS, and mapped its functional domains. We found KicGAS self-oligomerizes and binds to double stranded DNA cooperatively. This self-oligomerization is essential for its DNA binding and cGAS inhibition. Interestingly, KicGAS forms liquid droplets upon binding to DNA, which requires collective multivalent interactions with DNA mediated by both structured and disordered domains coordinated through the self-oligomerization of KicGAS. We also observed that KicGAS inhibits the DNA-induced phase separation and activation of cGAS. Our findings reveal a novel mechanism by which DNA viruses target the host protein phase separation for suppression of the host sensing of viral nucleic acids.


Asunto(s)
Herpesvirus Humano 8/genética , Interacciones Huésped-Patógeno/genética , Nucleotidiltransferasas/genética , Sarcoma de Kaposi/genética , Citosol/enzimología , Citosol/microbiología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/genética , ADN Viral/genética , Proteínas de Unión al ADN/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Evasión Inmune/efectos de los fármacos , Inmunidad Innata/genética , Nucleótidos Cíclicos/genética , Nucleotidiltransferasas/antagonistas & inhibidores , Sarcoma de Kaposi/tratamiento farmacológico , Sarcoma de Kaposi/virología , Proteínas Virales/genética
7.
Cell Rep ; 35(5): 109056, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33951431

RESUMEN

Herpesvirus infections shape the human natural killer (NK) cell compartment. While Epstein-Barr virus (EBV) expands immature NKG2A+ NK cells, human cytomegalovirus (CMV) drives accumulation of adaptive NKG2C+ NK cells. Kaposi sarcoma-associated herpesvirus (KSHV) is a close relative of EBV, and both are associated with lymphomas, including primary effusion lymphoma (PEL), which nearly always harbors both viruses. In this study, KSHV dual infection of mice with reconstituted human immune system components leads to the accumulation of CD56-CD16+CD38+CXCR6+ NK cells. CD56-CD16+ NK cells were also more frequently found in KSHV-seropositive Kenyan children. This NK cell subset is poorly cytotoxic against otherwise-NK-cell-susceptible and antibody-opsonized targets. Accordingly, NK cell depletion does not significantly alter KSHV infection in humanized mice. These data suggest that KSHV might escape NK-cell-mediated immune control by driving CD56-CD16+ NK cell differentiation.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 8/patogenicidad , Células Asesinas Naturales/inmunología , Animales , Diferenciación Celular , Humanos , Ratones
8.
J Med Virol ; 93(8): 5065-5075, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33942339

RESUMEN

Viral infection has been implicated in the pathogenesis of a plethora of human diseases. Although antiviral therapies effectively confront the viral spread and infection, how to completely eradicate the viral genome from infected cells remains a challenge. In this study, we demonstrated the reversible switching of primary cells between normal and malignant states by an oncogenic virus Kaposi's sarcoma-associated herpesvirus (KSHV) and CRISPR/Cas9-mediated targeting of a major viral latent protein. Primary cells can be transformed into malignant status by infection of KSHV, while elimination of the KSHV genome from latent KSHV-infected cells reverses KSHV-transformed primary cells back to a "normal state" by CRISPR/Cas-mediated knockout of viral major latent gene LANA. As a proof of concept, we demonstrated efficient elimination of KSHV episome in KSHV-associated primary effusion lymphoma cells resulting in the induction of apoptosis by liposome-encapsulated CRISPR/Cas9 ribonucleoprotein complexes (Lipo/Cas9-LANAsgRNA). Our work illustrates CRISPR/Cas as a promising technology for eliminating oncogenic viruses from persistently infected cells by taking advantage of the genetic differences between viral and cellular genomes. Compared to traditional antiviral therapy, our study offer an approach for antagonizing human oncogenic virus-related cancers by directly targeting as well as clearing viral genomes.


Asunto(s)
Antígenos Virales/genética , Sistemas CRISPR-Cas , Transformación Celular Neoplásica/genética , Herpesvirus Humano 8/genética , Proteínas Nucleares/genética , Virus Oncogénicos/genética , Animales , Antígenos Virales/metabolismo , Apoptosis , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Ciclo Celular , Proliferación Celular , Técnicas de Inactivación de Genes , Genoma Viral/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Linfoma de Efusión Primaria/patología , Células Madre Mesenquimatosas , Proteínas Nucleares/metabolismo , Virus Oncogénicos/patogenicidad , ARN Guía de Kinetoplastida/genética , Ratas , Latencia del Virus/genética
9.
Sci Rep ; 11(1): 7640, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33828146

RESUMEN

Human gammaherpesvirus 8 (HHV-8) consists of six major clades (A-F) based on the genetic sequence of the open reading frame (ORF)-K1. There are a few conflicting reports regarding the global distribution of the different HHV-8 genotypes. This study aimed to determine the global distribution of the different HHV-8 genotypes based on phylogenetic analysis of the ORF-K1 coding region using sequences published in the GenBank during 1997-2020 and construct a phylogenetic tree using the maximum likelihood algorithm with the GTR + I + G nucleotide substitution model. A total of 550 sequences from 38 countries/origins were analysed in this study. Genotypes A and C had similar global distributions and were prevalent in Africa and Europe. Genotype B was prevalent in Africa. Of the rare genotypes, genotype D was reported in East Asia and Oceania and genotype E in South America, while genotype F was prevalent in Africa. The highest genotypic diversity was reported in the American continent, with Brazil housing five HHV-8 genotypes (A, B, C, E, and F). In this study, we present update of the global distribution of HHV-8 genotypes, providing a basis for future epidemiological and evolutionary studies of HHV-8.


Asunto(s)
Infecciones por Herpesviridae/epidemiología , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/genética , ADN Viral/genética , Bases de Datos Genéticas , Gammaherpesvirinae/genética , Gammaherpesvirinae/patogenicidad , Variación Genética/genética , Genotipo , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/patogenicidad , Humanos , Sistemas de Lectura Abierta/genética , Filogenia , Sarcoma de Kaposi/epidemiología , Sarcoma de Kaposi/virología , Análisis de Secuencia de ADN/métodos , Proteínas Virales/genética
10.
Viruses ; 13(4)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33920978

RESUMEN

The oral cavity is often the first site where viruses interact with the human body. The oral epithelium is a major site of viral entry, replication and spread to other cell types, where chronic infection can be established. In addition, saliva has been shown as a primary route of person-to-person transmission for many viruses. From a clinical perspective, viral infection can lead to several oral manifestations, ranging from common intraoral lesions to tumors. Despite the clinical and biological relevance of initial oral infection, little is known about the mechanism of regulation of the viral life cycle in the oral cavity. Several viruses utilize host epigenetic machinery to promote their own life cycle. Importantly, viral hijacking of host chromatin-modifying enzymes can also lead to the dysregulation of host factors and in the case of oncogenic viruses may ultimately play a role in promoting tumorigenesis. Given the known roles of epigenetic regulation of viral infection, epigenetic-targeted antiviral therapy has been recently explored as a therapeutic option for chronic viral infection. In this review, we highlight three herpesviruses with known roles in oral infection, including herpes simplex virus type 1, Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus. We focus on the respective oral clinical manifestations of these viruses and their epigenetic regulation, with a specific emphasis on the viral life cycle in the oral epithelium.


Asunto(s)
Epigénesis Genética , Regulación Viral de la Expresión Génica , Herpesviridae/genética , Enfermedades de la Boca/virología , Saliva/virología , Replicación Viral/genética , Línea Celular , Herpesviridae/clasificación , Herpesviridae/patogenicidad , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidad , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Boca/patología , Boca/virología , Internalización del Virus
11.
J Med Virol ; 93(6): 3294-3299, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33415746

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) represents the etiological agent for several human malignancies, including Kaposi's Sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease (MCD), which develop mainly in immunocompromised patients. KSHV has established many strategies to hijack and thwart the host's immune responses, including through the use of extracellular vesicles (EVs). EVs represent a significant mode of intercellular communication as they carry a variety of molecules that can be delivered from cell-to-cell. EVs are now recognized as one of the major players in immune system development and function during both innate and adaptive immune responses. In the current mini-review, we summarize recent findings on how KSHV utilizes EVs to create favorable environments for viral spread and persistence while evading immune responses. We also discuss the limitations and unanswered questions in this field and the potential areas for related immunotherapies.


Asunto(s)
Vesículas Extracelulares/virología , Herpesvirus Humano 8/patogenicidad , Sarcoma de Kaposi/virología , Herpesvirus Humano 8/inmunología , Humanos , Evasión Inmune
12.
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
13.
Nat Commun ; 11(1): 6318, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33298918

RESUMEN

Primary effusion lymphoma (PEL) has a very poor prognosis. To evaluate the contributions of enhancers/promoters interactions to PEL cell growth and survival, here we produce H3K27ac HiChIP datasets in PEL cells. This allows us to generate the PEL enhancer connectome, which links enhancers and promoters in PEL genome-wide. We identify more than 8000 genomic interactions in each PEL cell line. By incorporating HiChIP data with H3K27ac ChIP-seq data, we identify interactions between enhancers/enhancers, enhancers/promoters, and promoters/promoters. HiChIP further links PEL super-enhancers to PEL dependency factors MYC, IRF4, MCL1, CCND2, MDM2, and CFLAR. CRISPR knock out of MEF2C and IRF4 significantly reduces MYC and IRF4 super-enhancer H3K27ac signal. Knock out also reduces MYC and IRF4 expression. CRISPRi perturbation of these super-enhancers by tethering transcription repressors to enhancers significantly reduces target gene expression and reduces PEL cell growth. These data provide insights into PEL molecular pathogenesis.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Redes Reguladoras de Genes , Linfoma de Efusión Primaria/genética , Regiones Promotoras Genéticas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Secuenciación de Inmunoprecipitación de Cromatina , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Herpesvirus Humano 8/patogenicidad , Histonas/genética , Humanos , Factores Reguladores del Interferón/genética , Linfoma de Efusión Primaria/patología , Linfoma de Efusión Primaria/virología , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética
14.
Front Immunol ; 11: 581419, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33013936

RESUMEN

Mice with reconstituted human immune systems can mount cell-mediated immune responses against the human tumor viruses Epstein Barr virus (EBV) and Kaposi sarcoma associated herpesvirus (KSHV). Primarily cytotoxic lymphocytes protect the vast majority of persistently infected carriers of these tumor viruses from the respective malignancies for life. Thus, EBV and KSHV infection can teach us how this potent immune control is induced, what phenotype and functions characterize the protective lymphocyte compartments and if similar immune responses could be induced by vaccination. This review will summarize similarities and differences between EBV and KSHV associated pathologies and their immune control in patients and mice with reconstituted human immune systems. Furthermore, it will high-light which aspects of the near perfect immune control can be modeled in the latter preclinical animal models and discuss their relevance for cancer immunology in general.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 8/inmunología , Animales , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/prevención & control , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 8/patogenicidad , Humanos , Inmunidad Innata , Leucocitos/inmunología , Ratones , Ratones SCID , Linfocitos T Citotóxicos/inmunología , Vacunación
15.
Biochem Biophys Res Commun ; 533(4): 1400-1405, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33092788

RESUMEN

The CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated gene 9) system is an RNA-guided, DNA editing method that has been widely used for gene editing, including human viruses. Kaposi's sarcoma-associated herpesvirus (KSHV/HHV8), following latent infection in human cells, can cause a variety of malignancies, such as Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman disease (MCD), with a high prevalence in immunocompromised patients. Of significant concern, the latent infection with KSHV has been shown to lead to increased resistance to antiviral therapies. MicroRNAs (miRNAs) are a set of non-coding, small RNA molecules that regulate protein-coding genes at the post-transcriptional and translational levels. KSHV has its miRNAs, most of which are expressed in latently infected cells and play a crucial role in maintaining KSHV latency. Notably, by regulating the expression of the downstream target genes in host cells, KSHV miRNAs can interact with the host environment to promote the development of KSHV-related diseases. Although CRISPR/Cas9 has been reported to edit KSHV protein-coding genes, there is no published literature on whether the CRISPR/Cas9 system can regulate the expression of KSHV miRNAs. In this study, we used CRISPR/Cas9 to inhibit the expression of KSHV miRNAs by directly editing the DNA sequences of individual KSHV miRNAs, or the promoter of clustered KHSV miRNAs, in latent KSHV-infected PEL cells. Our results show that CRISPR/Cas9 can ablate KSHV miRNAs expression, which in turn leads to the upregulation of viral lytic genes and alteration of host cellular gene expression. To the best of our knowledge, our study is the first reported demonstration of the CRISPR/Cas9 system editing KSHV miRNAs, further expanding the application of CRISPR/Cas9 as a novel antiviral strategy targeting KSHV latency.


Asunto(s)
Sistemas CRISPR-Cas , Herpesvirus Humano 8/genética , Infección Latente/genética , Linfoma de Efusión Primaria/virología , Edición Génica , Regulación Viral de la Expresión Génica , Genoma Viral , Herpesvirus Humano 8/patogenicidad , Interacciones Huésped-Patógeno/genética , Humanos , MicroARNs , Oxígeno/metabolismo , Medicina de Precisión/métodos , Sarcoma de Kaposi/virología
16.
mBio ; 11(4)2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843547

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) causes primary effusion lymphoma (PEL). The cellular transcription factor (TF) interferon (IFN) regulatory factor 4 (IRF4) is an essential oncogene in PEL, but its specific role in PEL and how KSHV deregulates IRF4 remain unknown. Here, we report that the KSHV latency protein viral interferon regulatory factor 3 (vIRF3) cooperates with IRF4 and cellular BATF (basic leucine zipper ATF-like TF) to drive a super-enhancer (SE)-mediated oncogenic transcriptional program in PEL. Chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-Seq) experiments demonstrated that IRF4, vIRF3, and BATF cooccupy the SEs of key survival genes, in a pattern that is distinct from those seen with other IRF4-driven malignancies. All three proteins cooperatively drive SE-mediated IRF4 overexpression. Inactivation of vIRF3 and, to a lesser extent, BATF phenocopies the gene expression changes and loss of cellular viability observed upon inactivation of IRF4. In sum, this work suggests that KSHV vIRF3 and cellular IRF4 and BATF cooperate as oncogenic transcription factors on SEs to promote cellular survival and proliferation in KSHV-associated lymphomas.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) causes the aggressive disease primary effusion lymphoma (PEL). Here, we show that a viral transcription factor (vIRF3) cooperates with the cellular transcription factor IRF4 to control an oncogenic gene expression program in PEL cells. These proteins promote KSHV-mediated B cell transformation by activating the expression of prosurvival genes through super-enhancers. Our report thus demonstrates that this DNA tumor virus encodes a transcription factor that functions with cellular IRF4 to drive oncogenic transcriptional reprogramming.


Asunto(s)
Expresión Génica , Herpesvirus Humano 8/patogenicidad , Linfoma de Efusión Primaria/genética , Linfoma de Efusión Primaria/virología , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virología , Línea Celular Tumoral , Humanos , Factores Reguladores del Interferón/genética , Proteínas Virales/genética , Latencia del Virus
17.
Proc Natl Acad Sci U S A ; 117(35): 21618-21627, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817485

RESUMEN

Enhancers play indispensable roles in cell proliferation and survival through spatiotemporally regulating gene transcription. Active enhancers and superenhancers often produce noncoding enhancer RNAs (eRNAs) that precisely control RNA polymerase II activity. Kaposi's sarcoma-associated herpesvirus (KSHV) is a human oncogenic gamma-2 herpesvirus that causes Kaposi's sarcoma and primary effusion lymphoma (PEL). It is well characterized that KSHV utilizes host epigenetic machineries to control the switch between two lifecycles, latency and lytic replication. However, how KSHV impacts host epigenome at different stages of viral lifecycle is not well understood. Using global run-on sequencing (GRO-seq) and chromatin-immunoprecipitation sequencing (ChIP-seq), we profiled the dynamics of host transcriptional regulatory elements during latency and lytic replication of KSHV-infected PEL cells. This revealed that a number of critical host genes for KSHV latency, including MYC proto-oncogene, were under the control of superenhancers whose activities were globally repressed upon viral reactivation. The eRNA-expressing MYC superenhancers were located downstream of the MYC gene in KSHV-infected PELs and played a key role in MYC expression. RNAi-mediated depletion or dCas9-KRAB CRISPR inhibition of eRNA expression significantly reduced MYC mRNA level in PELs, as did the treatment of an epigenomic drug that globally blocks superenhancer function. Finally, while cellular IRF4 acted upon eRNA expression and superenhancer function for MYC expression during latency, KSHV viral IRF4 repressed cellular IRF4 expression, decreasing MYC expression and thereby, facilitating lytic replication. These results indicate that KSHV acts as an epigenomic driver that modifies host epigenomic status upon reactivation by effectively regulating host enhancer function.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/genética , Linfoma de Efusión Primaria/genética , Línea Celular , Epigenómica/métodos , Genes myc/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Proteínas Inmediatas-Precoces/genética , Linfoma de Efusión Primaria/metabolismo , Linfoma de Efusión Primaria/virología , Proteínas Nucleares/metabolismo , Proto-Oncogenes Mas , ARN/metabolismo , Sarcoma de Kaposi/virología , Transactivadores/metabolismo , Transcripción Genética/genética , Proteínas Virales/metabolismo , Activación Viral/genética , Latencia del Virus/genética , Replicación Viral/genética
18.
Cells ; 9(9)2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32854341

RESUMEN

RNA-binding proteins, particularly splicing factors, localize to sub-nuclear domains termed nuclear speckles. During certain viral infections, as the nucleus fills up with replicating virus compartments, host cell chromatin distribution changes, ending up condensed at the nuclear periphery. In this study we wished to determine the fate of nucleoplasmic RNA-binding proteins and nuclear speckles during the lytic cycle of the Kaposi's sarcoma associated herpesvirus (KSHV). We found that nuclear speckles became fewer and dramatically larger, localizing at the nuclear periphery, adjacent to the marginalized chromatin. Enlarged nuclear speckles contained splicing factors, whereas other proteins were nucleoplasmically dispersed. Polyadenylated RNA, typically found in nuclear speckles under regular conditions, was also found in foci separated from nuclear speckles in infected cells. Poly(A) foci did not contain lncRNAs known to colocalize with nuclear speckles but contained the poly(A)-binding protein PABPN1. Examination of the localization of spliced viral RNAs revealed that some spliced transcripts could be detected within the nuclear speckles. Since splicing is required for the maturation of certain KSHV transcripts, we suggest that the infected cell does not dismantle nuclear speckles but rearranges their components at the nuclear periphery to possibly serve in splicing and transport of viral RNAs into the cytoplasm.


Asunto(s)
Núcleo Celular/virología , Herpesvirus Humano 8/patogenicidad , Sarcoma de Kaposi/genética , Humanos , Proteínas de Unión al ARN/metabolismo
19.
Virology ; 549: 5-12, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32777727

RESUMEN

Kaposi's sarcoma (KS) tends to occur in injured or inflamed sites of the body, which is described as the "Koebner phenomenon". KS is also unique in its extraordinary angio-hyperplastic inflammatory phenotype. Recently, evidence has accrued indicating that KS may derive from KSHV-infected mesenchymal stem cells (MSCs), which possess enhanced migration and homing ability. Inspired by these findings, we hypothesized that KS may arise from KSHV-infected MSCs that chemotactically migrate to preexisting inflammatory or injured sites. Here we report that KSHV infection of human MSCs significantly up-regulated expression of several chemokine receptors and enhanced cell migration ability in vitro. Furthermore, using a wound mouse model, we demonstrated that KSHV infection dramatically promotes MSCs migrating and settling in the wound sites. In addition, two mice in the KSHV-infected group showed purpura and tumors with KS-like features. Taken together, KSHV-enhanced MSC migration ability and inflammatory microenvironment play crucial roles in KS development.


Asunto(s)
Herpesvirus Humano 8/patogenicidad , Receptores CCR1/genética , Receptores CCR3/genética , Receptores CCR4/genética , Sarcoma de Kaposi/genética , Heridas no Penetrantes/virología , Animales , Movimiento Celular , Modelos Animales de Enfermedad , Oído , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Herpesvirus Humano 8/crecimiento & desarrollo , Interacciones Huésped-Patógeno/genética , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ligamento Periodontal/citología , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores CCR1/antagonistas & inhibidores , Receptores CCR1/metabolismo , Receptores CCR3/antagonistas & inhibidores , Receptores CCR3/metabolismo , Receptores CCR4/antagonistas & inhibidores , Receptores CCR4/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/virología , Transducción de Señal , Células Madre/metabolismo , Células Madre/patología , Células Madre/virología , Heridas no Penetrantes/patología , Proteína Fluorescente Roja
20.
Nat Commun ; 11(1): 3849, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32737300

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr Virus (EBV) establish life-long infections and are associated with malignancies. Striking geographic variation in incidence and the fact that virus alone is insufficient to cause disease, suggests other co-factors are involved. Here we present epidemiological analysis and genome-wide association study (GWAS) in 4365 individuals from an African population cohort, to assess the influence of host genetic and non-genetic factors on virus antibody responses. EBV/KSHV co-infection (OR = 5.71(1.58-7.12)), HIV positivity (OR = 2.22(1.32-3.73)) and living in a more rural area (OR = 1.38(1.01-1.89)) are strongly associated with immunogenicity. GWAS reveals associations with KSHV antibody response in the HLA-B/C region (p = 6.64 × 10-09). For EBV, associations are identified for VCA (rs71542439, p = 1.15 × 10-12). Human leucocyte antigen (HLA) and trans-ancestry fine-mapping substantiate that distinct variants in HLA-DQA1 (p = 5.24 × 10-44) are driving associations for EBNA-1 in Africa. This study highlights complex interactions between KSHV and EBV, in addition to distinct genetic architectures resulting in important differences in pathogenesis and transmission.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Resistencia a la Enfermedad/genética , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Henipavirus/genética , Interacciones Huésped-Patógeno/genética , Sarcoma de Kaposi/genética , Adolescente , Adulto , Antígenos Virales/genética , Antígenos Virales/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Coinfección , Infecciones por Virus de Epstein-Barr/epidemiología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/genética , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Femenino , Expresión Génica , Estudio de Asociación del Genoma Completo , VIH/genética , VIH/inmunología , VIH/patogenicidad , Cadenas alfa de HLA-DQ/genética , Cadenas alfa de HLA-DQ/inmunología , Infecciones por Henipavirus/epidemiología , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/virología , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Herpesvirus Humano 8/patogenicidad , Interacciones Huésped-Patógeno/inmunología , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Población Rural , Sarcoma de Kaposi/epidemiología , Sarcoma de Kaposi/inmunología , Sarcoma de Kaposi/virología , Uganda/epidemiología , Población Urbana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...