Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(4): 1814-1829, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38180827

RESUMEN

To establish lifelong, latent infection, herpesviruses circularize their linear, double-stranded, DNA genomes through an unknown mechanism. Kaposi's sarcoma (KS) herpesvirus (KSHV), a gamma herpesvirus, is tightly linked with KS, primary effusion lymphoma, and multicentric Castleman's disease. KSHV persists in latently infected cells as a multi-copy, extrachromosomal episome. Here, we show the KSHV genome rapidly circularizes following infection, and viral protein expression is unnecessary for this process. The DNA damage response (DDR) kinases, ATM and DNA-PKcs, each exert roles, and absence of both severely compromises circularization and latency. These deficiencies were rescued by expression of ATM and DNA-PKcs, but not catalytically inactive mutants. In contrast, γH2AX did not function in KSHV circularization. The linear viral genomic ends resemble a DNA double strand break, and non-homologous DNA end joining (NHEJ) and homologous recombination (HR) reporters indicate both NHEJ and HR contribute to KSHV circularization. Last, we show, similar to KSHV, ATM and DNA-PKcs have roles in circularization of the alpha herpesvirus, herpes simplex virus-1 (HSV-1), while γH2AX does not. Therefore, the DDR mediates KSHV and HSV-1 circularization. This strategy may serve as a general herpesvirus mechanism to initiate latency, and its disruption may provide new opportunities for prevention of herpesvirus disease.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/genética , Latencia del Virus/genética , ADN , Reparación del ADN
2.
PLoS Pathog ; 20(1): e1011907, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38232124

RESUMEN

Kaposi's sarcoma herpesvirus (KSHV) is a leading cause of malignancy in AIDS and current therapies are limited. Like all herpesviruses, KSHV infection can be latent or lytic. KSHV latency-associated nuclear antigen (LANA) is essential for viral genome persistence during latent infection. LANA also maintains latency by antagonizing expression and function of the KSHV lytic switch protein, RTA. Here, we find LANA null KSHV is not capable of lytic replication, indicating a requirement for LANA. While LANA promoted both lytic and latent gene expression in cells partially permissive for lytic infection, it repressed expression in non-permissive cells. Importantly, forced RTA expression in non-permissive cells led to induction of lytic infection and LANA switched to promote, rather than repress, most lytic viral gene expression. When basal viral gene expression levels were high, LANA promoted expression, but repressed expression at low basal levels unless RTA expression was forcibly induced. LANA's effects were broad, but virus gene specific, extending to an engineered, recombinant viral GFP under control of host EF1α promoter, but not to host EF1α. Together, these results demonstrate that, in addition to its essential role in genome maintenance, LANA broadly regulates viral gene expression, and is required for high levels of lytic gene expression during lytic infection. Strategies that target LANA are expected to abolish KSHV infection.


Asunto(s)
Herpesvirus Humano 8 , Proteínas Nucleares , Sarcoma de Kaposi , Humanos , Herpesvirus Humano 8/fisiología , Latencia del Virus/genética , Antígenos Virales/genética , Antígenos Virales/metabolismo , Expresión Génica , Regulación Viral de la Expresión Génica , Replicación Viral
3.
Medicine (Baltimore) ; 102(48): e36417, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38050198

RESUMEN

Little information is available regarding incidence and severity of pulmonary embolism (PE) across the periods of ancestral strain, Alpha, Delta, and Omicron variants. The aim of this study is to investigate the incidence and severity of PE over the dominant periods of ancestral strain and Alpha, Delta, and Omicron variants. We hypothesized that the incidence and the severity by proximity of PE in patients with the newer variants and vaccination would be decreased compared with those in ancestral and earlier variants. Patients with COVID-19 diagnosis between March 2020 and February 2022 and computed tomography pulmonary angiogram performed within a 6-week window around the diagnosis (-2 to +4 weeks) were studied retrospectively. The primary endpoints were the associations of the incidence and location of PE with the ancestral strain and each variant. Of the 720 coronavirus disease 2019 patients with computed tomography pulmonary angiogram (58.6 ± 17.2 years; 374 females), PE was diagnosed among 42/358 (12%) during the ancestral strain period, 5/60 (8%) during the Alpha variant period, 16/152 (11%) during the Delta variant period, and 13/150 (9%) during the Omicron variant period. The most proximal PE (ancestral strain vs variants) was located in the main/lobar arteries (31% vs 6%-40%), in the segmental arteries (52% vs 60%-75%), and in the subsegmental arteries (17% vs 0%-19%). There was no significant difference in both the incidence and location of PE across the periods, confirmed by multivariable logistic regression models. In summary, the incidence and severity of PE did not significantly differ across the periods of ancestral strain and Alpha, Delta, and Omicron variants.


Asunto(s)
COVID-19 , Embolia Pulmonar , Femenino , Humanos , Prueba de COVID-19 , Incidencia , Estudios Retrospectivos , COVID-19/epidemiología , SARS-CoV-2 , Embolia Pulmonar/diagnóstico por imagen , Embolia Pulmonar/epidemiología , Arteria Pulmonar
4.
Cell Rep ; 42(7): 112767, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37440412

RESUMEN

Kaposi's sarcoma herpesvirus (KSHV) establishes lifelong infection and persists in latently infected B cells. Paradoxically, in vitro B cell infection is inefficient, and cells rapidly die, suggesting the absence of necessary factor(s). KSHV epidemiology unexpectedly mirrors that of malaria and certain helminthic infections, while other herpesviruses are ubiquitous. Elevated circulating monocytes are common in these parasitic infections. Here, we show that KSHV infection of monocytes or M-CSF-differentiated (M2) macrophages is highly efficient. Proteomic analyses demonstrate that infection induces macrophage production of B cell chemoattractants and activating factor. We find that KSHV acts with monocytes or M2 macrophages to stimulate B cell survival, proliferation, and plasmablast differentiation. Further, macrophages drive infected plasma cell differentiation and long-term viral latency. In Kenya, where KSHV is endemic, we find elevated monocyte levels in children with malaria. These findings demonstrate a role for mononuclear phagocytes in KSHV B cell latency and suggest that mononuclear phagocyte abundance may underlie KSHV's geographic disparity.


Asunto(s)
Herpesvirus Humano 8 , Niño , Humanos , Proteómica , Linfocitos B , Macrófagos , Monocitos , Latencia del Virus
5.
Eur J Radiol Open ; 10: 100483, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36883046

RESUMEN

Purpose: To investigate the association of the maximal severity of pneumonia on CT scans obtained within 6-week of diagnosis with the subsequent development of post-COVID-19 lung abnormalities (Co-LA). Methods: COVID-19 patients diagnosed at our hospital between March 2020 and September 2021 were studied retrospectively. The patients were included if they had (1) at least one chest CT scan available within 6-week of diagnosis; and (2) at least one follow-up chest CT scan available ≥ 6 months after diagnosis, which were evaluated by two independent radiologists. Pneumonia Severity Categories were assigned on CT at diagnosis according to the CT patterns of pneumonia and extent as: 1) no pneumonia (Estimated Extent, 0%); 2) non-extensive pneumonia (GGO and OP, <40%); and 3) extensive pneumonia (extensive OP and DAD, >40%). Co-LA on follow-up CT scans, categorized using a 3-point Co-LA Score (0, No Co-LA; 1, Indeterminate Co-LA; and 2, Co-LA). Results: Out of 132 patients, 42 patients (32%) developed Co-LA on their follow-up CT scans 6-24 months post diagnosis. The severity of COVID-19 pneumonia was associated with Co-LA: In 47 patients with extensive pneumonia, 33 patients (70%) developed Co-LA, of whom 18 (55%) developed fibrotic Co-LA. In 52 with non-extensive pneumonia, 9 (17%) developed Co-LA: In 33 with no pneumonia, none (0%) developed Co-LA. Conclusions: Higher severity of pneumonia at diagnosis was associated with the increased risk of development of Co-LA after 6-24 months of SARS-CoV-2 infection.

8.
Eur J Radiol Open ; 9: 100456, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36386765

RESUMEN

Purpose: To investigate the effect of vaccinations and boosters on the severity of COVID-19 pneumonia on CT scans during the period of Delta and Omicron variants. Methods: Retrospectively studied were 303 patients diagnosed with COVID-19 between July 2021 and February 2022, who had obtained at least one CT scan within 6 weeks around the COVID-19 diagnosis (-2 to +4 weeks). The severity of pneumonia was evaluated with a 6-point scale Pneumonia Score. The association between demographic and clinical data and vaccination status (booster/additional vaccination, complete vaccination and un-vaccination) and the difference between Pneumonia Scores by vaccination status were investigated. Results: Of 303 patients (59.4 ± 16.3 years; 178 females), 62 (20 %) were in the booster/additional vaccination group, 117 (39 %) in the complete vaccination group, and 124 (41 %) in the unvaccinated group. Interobserver agreement of the Pneumonia Score was high (weighted kappa score = 0.875). Patients in the booster/additionally vaccinated group tended to be older (P = 0.0085) and have more underlying comorbidities (P < 0.0001), and the Pneumonia Scores were lower in the booster/additionally vaccinated [median 2 (IQR 0-4)] and completely vaccinated groups [median 3 (IQR 1-4)] than those in the unvaccinated group [median 4 (IQR 2-4)], respectively (P < 0.0001 and P < 0.0001, respectively). A multivariable linear analysis adjusted for confounding factors confirmed the difference. Conclusion: Vaccinated patients, with or without booster/additional vaccination, had milder COVID-19 pneumonia on CT scans than unvaccinated patients during the period of Delta and Omicron variants. This study supports the efficacy of the vaccine against COVID-19 from a radiological perspective.

9.
Nucleic Acids Res ; 49(22): 12895-12911, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34850113

RESUMEN

Mixed lineage leukemia 1 (MLL1) is a histone methyltransferase. Kaposi's sarcoma-associated herpesvirus (KSHV) is a leading cause of malignancy in AIDS. KSHV latently infects tumor cells and its genome is decorated with epigenetic marks. Here, we show that KSHV latency-associated nuclear antigen (LANA) recruits MLL1 to viral DNA where it establishes H3K4me3 modifications at the extensive KSHV terminal repeat elements during primary infection. LANA interacts with MLL1 complex members, including WDR5, integrates into the MLL1 complex, and regulates MLL1 activity. We describe the 1.5-Å crystal structure of N-terminal LANA peptide complexed with MLL1 complex member WDR5, which reveals a potential regulatory mechanism. Disruption of MLL1 expression rendered KSHV latency establishment highly deficient. This deficiency was rescued by MLL1 but not by catalytically inactive MLL1. Therefore, MLL1 is LANA regulable and exerts a central role in virus infection. These results suggest broad potential for MLL1 regulation, including by non-host factors.


Asunto(s)
Antígenos Virales/genética , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , N-Metiltransferasa de Histona-Lisina/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Nucleares/genética , Sarcoma de Kaposi/genética , Latencia del Virus/genética , Antígenos Virales/química , Antígenos Virales/metabolismo , Línea Celular Tumoral , Cristalografía por Rayos X , ADN Viral/genética , ADN Viral/metabolismo , Técnicas de Silenciamiento del Gen , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/fisiología , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/química , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Unión Proteica , Conformación Proteica , Sarcoma de Kaposi/virología
10.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34725152

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) causes the endothelial tumor KS, a leading cause of morbidity and mortality in sub-Saharan Africa. KSHV-encoded microRNAs (miRNAs) are known to play an important role in viral oncogenesis; however, the role of host miRNAs in KS tumorigenesis remains largely unknown. Here, high-throughput small-RNA sequencing of the cellular transcriptome in a KS xenograft model revealed miR-127-3p as one of the most significantly down-regulated miRNAs, which we validated in KS patient tissues. We show that restoration of miR-127-3p suppresses KSHV-driven cellular transformation and proliferation and induces G1 cell cycle arrest by directly targeting the oncogene SKP2. This miR-127-3p-induced G1 arrest is rescued by disrupting the miR-127-3p target site in SKP2 messenger RNA (mRNA) using gene editing. Mechanistically, miR-127-3p-mediated SKP2 repression elevates cyclin-dependent kinase (CDK) inhibitor p21Cip1 and down-regulates cyclin E, cyclin A, and CDK2, leading to activation of the RB protein tumor suppressor pathway and suppression of the transcriptional activities of E2F and Myc, key oncoprotein transcription factors crucial for KSHV tumorigenesis. Consequently, metabolomics analysis during miR-127-3p-induced cell cycle arrest revealed significant depletion of dNTP pools, consistent with RB-mediated repression of key dNTP biosynthesis enzymes. Furthermore, miR-127-3p reconstitution in a KS xenograft mouse model suppresses KSHV-positive tumor growth by targeting SKP2 in vivo. These findings identify a previously unrecognized tumor suppressor function for miR-127-3p in KS and demonstrate that the miR-127-3p/SKP2 axis is a viable therapeutic strategy for KS.


Asunto(s)
Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Sarcoma de Kaposi/metabolismo , Animales , Carcinogénesis , Femenino , Herpesvirus Humano 8/fisiología , Humanos , Ratones Desnudos , Sarcoma de Kaposi/virología
11.
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
12.
Proc Natl Acad Sci U S A ; 117(36): 22443-22451, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32820070

RESUMEN

Viruses modulate biochemical cellular pathways to permit infection. A recently described mechanism mediates selective protein interactions between acidic domain readers and unacetylated, lysine-rich regions, opposite of bromodomain function. Kaposi´s sarcoma (KS)-associated herpesvirus (KSHV) is tightly linked with KS, primary effusion lymphoma, and multicentric Castleman's disease. KSHV latently infects cells, and its genome persists as a multicopy, extrachromosomal episome. During latency, KSHV expresses a small subset of genes, including the latency-associated nuclear antigen (LANA), which mediates viral episome persistence. Here we show that LANA contains two tandem, partially overlapping, acidic domain sequences homologous to the SET oncoprotein acidic domain reader. This domain selectively interacts with unacetylated p53, as evidenced by reduced LANA interaction after overexpression of CBP, which acetylates p53, or with an acetylation mimicking carboxyl-terminal domain p53 mutant. Conversely, the interaction of LANA with an acetylation-deficient p53 mutant is enhanced. Significantly, KSHV LANA mutants lacking the acidic domain reader sequence are deficient for establishment of latency and persistent infection. This deficiency was confirmed under physiological conditions, on infection of mice with a murine gammaherpesvirus 68 chimera expressing LANA, where the virus was highly deficient in establishing latent infection in germinal center B cells. Therefore, LANA's acidic domain reader is critical for viral latency. These results implicate an acetylation-dependent mechanism mediating KSHV persistence and expand the role of acidic domain readers.


Asunto(s)
Antígenos Virales/genética , Antígenos Virales/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Acetilación , Animales , Antígenos Virales/química , ADN Viral/genética , Femenino , Células HEK293 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/química , Plásmidos/genética
13.
J Mol Diagn ; 22(4): 476-487, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32068070

RESUMEN

Precision cancer medicine aims to classify tumors by site, histology, and molecular testing to determine an individualized profile of cancer alterations. Viruses are a major contributor to oncogenesis, causing 12% to 20% of all human cancers. Several viruses are causal of specific types of cancer, promoting dysregulation of signaling pathways and resulting in carcinogenesis. In addition, integration of viral DNA into the host (human) genome is a hallmark of some viral species. Tests for the presence of viral infection used in the clinical setting most often use quantitative PCR or immunohistochemical staining. Both approaches have limitations and need to be interpreted/scored appropriately. In some cases, results are not binary (virus present/absent), and it is unclear what to do with a weakly or partially positive result. In addition, viral testing of cancers is performed separately from tests to detect human genomic alterations and can thus be time-consuming and use limited valuable specimen. We present a hybrid-capture and massively parallel sequencing approach to detect viral infection that is integrated with targeted genomic analysis to provide a more complete tumor profile from a single sample.


Asunto(s)
Genoma Humano , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Neoplasias/diagnóstico , Neoplasias/etiología , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/virología , Transformación Celular Viral , Biología Computacional/métodos , Genoma Viral , Genómica/métodos , Genómica/normas , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Filogenia , Polimorfismo de Nucleótido Simple , Medicina de Precisión/métodos , Sensibilidad y Especificidad , Infecciones Tumorales por Virus/diagnóstico , Integración Viral
14.
BMJ Case Rep ; 12(9)2019 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-31570344

RESUMEN

A 37-year-old woman with a history of inflammatory bowel disease on mercaptopurine presented with a week of recurrent fever, headache, myalgias and mildly elevated serum transaminases and leucopenia. Her workup revealed primary cytomegalovirus (CMV) infection with atypical lymphocytosis, elevated viral load, positive IgM and negative IgG. Two weeks after her initial presentation, she developed odynophagia and diarrhoea prompting endoscopic evaluation with biopsies, which demonstrated CMV disease of the gastrointestinal tract. Her fever and systemic symptoms improved rapidly with initiation of intravenous ganciclovir. She was transitioned to and maintained on oral valganciclovir until two and half months after discharge when her symptoms and lab abnormalities had fully subsided.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Citomegalovirus/diagnóstico , Ganciclovir/uso terapéutico , Enfermedades Gastrointestinales/virología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Infecciones Oportunistas/virología , Adulto , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/inmunología , Femenino , Fiebre , Enfermedades Gastrointestinales/tratamiento farmacológico , Enfermedades Gastrointestinales/inmunología , Cefalea , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Mialgia , Infecciones Oportunistas/tratamiento farmacológico , Infecciones Oportunistas/inmunología , Resultado del Tratamiento , Carga Viral
15.
PLoS Pathog ; 15(2): e1007628, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30811506

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) latently infects host cells and establishes lifelong persistence as an extra-chromosomal episome in the nucleus. To persist in proliferating cells, the viral genome typically replicates once per cell cycle and is distributed into daughter cells. This process involves host machinery utilized by KSHV, however the underlying mechanisms are not fully elucidated. In present study, we found that N-Myc downstream regulated gene 1 (NDRG1), a cellular gene known to be non-detectable in primary B cells and endothelial cells which are the major cell types for KSHV infection in vivo, was highly upregulated by KSHV in these cells. We further demonstrated that the high expression of NDRG1 was regulated by latency-associated nuclear antigen (LANA), the major viral latent protein which tethers the viral genome to host chromosome and plays an essential role in viral genome maintenance. Surprisingly, knockdown of NDRG1 in KSHV latently infected cells resulted in a significant decrease of viral genome copy number in these cells. Interestingly, NDRG1 can directly interact with proliferating cell nuclear antigen (PCNA), a cellular protein which functions as a DNA polymerase clamp during DNA replication. Intriguingly, we found that NDRG1 forms a complex with LANA and PCNA and serves as a scaffold protein bridging these two proteins. We further demonstrated that NDRG1 is critical for mediating LANA to recruit PCNA onto terminal repeat (TR) of KSHV genome, and facilitates viral DNA replication and episome persistence. Taken together, our findings suggest that NDRG1 plays an important role in KSHV viral genome replication, and provide new clues for understanding of KSHV persistence.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Herpesvirus Humano 8/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Adulto , Antígenos Virales/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Núcleo Celular/metabolismo , Replicación del ADN , ADN Viral/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Genoma Viral , Células HEK293 , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteínas Nucleares/metabolismo , Plásmidos/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Regulación hacia Arriba , Latencia del Virus , Replicación Viral
16.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626680

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA) is a 1,162-amino-acid protein that mediates episome persistence of viral genomes. LANA binds the KSHV terminal-repeat (TR) sequence through its carboxy-terminal domain to mediate DNA replication. LANA simultaneously binds mitotic chromosomes and TR DNA to segregate virus genomes to daughter cell nuclei. Amino-terminal LANA attaches to chromosomes by binding histones H2A/H2B, and carboxy-terminal LANA contributes to mitotic-chromosome binding. Although amino- and carboxy-terminal LANA are essential for episome persistence, they are not sufficient, since deletion of all internal LANA sequence renders LANA highly deficient for episome maintenance. Internal LANA sequence upstream of the internal repeat elements contributes to episome segregation and persistence. Here, we investigate this region with a panel of LANA deletion mutants. Mutants retained the ability to associate with mitotic chromosomes and bind TR DNA. In contrast to prior results, deletion of most of this sequence did not reduce LANA's ability to mediate DNA replication. Deletions of upstream sequence within the region compromised segregation of TR DNA to daughter cells, as assessed by retention of green fluorescent protein (GFP) expression from a replication-deficient TR plasmid. However, deletion of this upstream sequence did not reduce episome maintenance. In contrast, deletions that included an 80-amino-acid sequence immediately downstream resulted in highly deficient episome persistence. LANA with this downstream sequence deleted maintained the ability to replicate and segregate TR DNA, suggesting a unique role for the residues. Therefore, this work identifies adjacent LANA regions with distinct roles in episome segregation and persistence.IMPORTANCE KSHV LANA mediates episomal persistence of viral genomes. LANA binds the KSHV terminal-repeat (TR) sequence to mediate DNA replication and tethers KSHV DNA to mitotic chromosomes to segregate genomes to daughter cell nuclei. Here, we investigate LANA sequence upstream of the internal repeat elements that contributes to episome segregation and persistence. Mutants with deletions within this sequence maintained the ability to bind mitotic chromosomes or bind and replicate TR DNA. Deletion of upstream sequence within the region reduced segregation of TR DNA to daughter cells, but not episome maintenance. In contrast, mutants with deletions of 80 amino acids immediately downstream were highly deficient for episome persistence yet maintained the ability to replicate and segregate TR DNA, the two principal components of episome persistence, suggesting another role for the residues. In summary, this work identifies adjacent LANA sequence with distinct roles in episome segregation and persistence.


Asunto(s)
Antígenos Virales/genética , Herpesvirus Humano 8/genética , Proteínas Nucleares/genética , Plásmidos/genética , Sarcoma de Kaposi/virología , Antígenos Nucleares/genética , Línea Celular , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/virología , Cromosomas/genética , Replicación del ADN/genética , ADN Viral/genética , Genoma Viral/genética , Células HEK293 , Humanos , Mitosis/genética , Secuencias Repetidas Terminales/genética , Proteínas Virales/genética , Latencia del Virus/genética , Replicación Viral/genética
17.
J Virol ; 92(21)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30111565

RESUMEN

The latency-associated nuclear antigen from Kaposi's sarcoma-associated herpesvirus (KSHV), kLANA, and its homolog from the murid herpesvirus 4 (MuHV-4), mLANA, are essential for viral latency. kLANA is nearly four times the size of mLANA, mainly due to an extensive central repeat region that is absent in mLANA. Both proteins harbor a C-terminal DNA binding domain (DBD). The DBD binds the terminal repeat (TR) DNA sequences of the viral genome to mediate persistence. Despite structural conservation, the kLANA and mLANA DBDs differ in sequence and mode of oligomerization. kLANA DBD oligomers are flexible and bent, while mLANA DBD oligomers bind DNA in a rigid, linear conformation. We previously reported that kLANA and mLANA acted reciprocally on TR sequences. Furthermore, a MuHV-4 expressing kLANA instead of mLANA (v-kLANA) established latency in mice, albeit at a lower magnitude than the wild-type (WT) virus. Here, we asked if kLANA can accommodate the mLANA DBD and generated a fusion protein which contains kLANA but with the mLANA C-terminal region in place of that of kLANA. We report a recombinant MuHV-4 (v-KM) encoding this LANA fusion protein instead of mLANA. The fusion protein was expressed in lytic infection in vitro and assembled nuclear LANA dots in infected splenocytes. Results demonstrated that kLANA functionally accommodated mLANA's mode of DNA binding, allowing MuHV-4 chimeric virus to establish latency in vivo Notably, v-KM established latency in germinal center B cells more efficiently than did v-kLANA, although levels were reduced compared to WT MuHV-4.IMPORTANCE KSHV is a human oncogenic virus for which there is no tractable, immunocompetent animal model of infection. MuHV-4, a related rodent gammaherpesvirus, enables pathogenesis studies in mice. In latency, both viruses persist as extrachromosomal, circular genomes (episomes). LANA proteins encoded by KSHV (kLANA) and MuHV-4 (mLANA) contain a C-terminal DNA binding domain (DBD) that acts on the virus terminal repeats to enable episome persistence. mLANA is a smaller protein than kLANA. Their DBDs are structurally conserved but differ strikingly in the conformation of DNA binding. We report a recombinant, chimeric MuHV-4 which contains kLANA in place of mLANA, but in which the DBD is replaced with that of mLANA. Results showed that kLANA functionally accommodated mLANA's mode of DNA binding. In fact, the new chimeric virus established latency in vivo more efficiently than MuHV-4 expressing full-length kLANA.


Asunto(s)
Antígenos Virales/metabolismo , ADN Viral/metabolismo , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 8/genética , Proteínas Nucleares/metabolismo , Rhadinovirus/genética , Secuencias Repetidas Terminales/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Línea Celular , ADN Viral/genética , Genoma Viral/genética , Ratones , Latencia del Virus/genética
18.
PLoS Pathog ; 13(9): e1006555, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28910389

RESUMEN

Many pathogens, including Kaposi's sarcoma herpesvirus (KSHV), lack tractable small animal models. KSHV persists as a multi-copy, nuclear episome in latently infected cells. KSHV latency-associated nuclear antigen (kLANA) binds viral terminal repeat (kTR) DNA to mediate episome persistence. Model pathogen murine gammaherpesvirus 68 (MHV68) mLANA acts analogously on mTR DNA. kLANA and mLANA differ substantially in size and kTR and mTR show little sequence conservation. Here, we find kLANA and mLANA act reciprocally to mediate episome persistence of TR DNA. Further, kLANA rescued mLANA deficient MHV68, enabling a chimeric virus to establish latent infection in vivo in germinal center B cells. The level of chimeric virus in vivo latency was moderately reduced compared to WT infection, but WT or chimeric MHV68 infected cells had similar viral genome copy numbers as assessed by immunofluorescence of LANA intranuclear dots or qPCR. Thus, despite more than 60 Ma of evolutionary divergence, mLANA and kLANA act reciprocally on TR DNA, and kLANA functionally substitutes for mLANA, allowing kLANA investigation in vivo. Analogous chimeras may allow in vivo investigation of genes of other human pathogens.


Asunto(s)
Antígenos Virales/metabolismo , ADN Viral/genética , Genoma Viral/genética , Centro Germinal/metabolismo , Herpesvirus Humano 8 , Proteínas Nucleares/metabolismo , Plásmidos/metabolismo , Sarcoma de Kaposi/metabolismo , Latencia del Virus/genética , Animales , Antígenos Virales/genética , Linfocitos B/metabolismo , Linfocitos B/virología , Ratones , Proteínas Nucleares/genética , Plásmidos/genética , Sarcoma de Kaposi/virología
19.
J Clin Invest ; 127(6): 2066-2080, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28504647

RESUMEN

Primary effusion lymphoma (PEL) is a largely incurable malignancy of B cell origin with plasmacytic differentiation. Here, we report the identification of a highly effective inhibitor of PEL. This compound, 6-ethylthioinosine (6-ETI), is a nucleoside analog with toxicity to PEL in vitro and in vivo, but not to other lymphoma cell lines tested. We developed and performed resistome analysis, an unbiased approach based on RNA sequencing of resistant subclones, to discover the molecular mechanisms of sensitivity. We found different adenosine kinase-inactivating (ADK-inactivating) alterations in all resistant clones and determined that ADK is required to phosphorylate and activate 6-ETI. Further, we observed that 6-ETI induces ATP depletion and cell death accompanied by S phase arrest and DNA damage only in ADK-expressing cells. Immunohistochemistry for ADK served as a biomarker approach to identify 6-ETI-sensitive tumors, which we documented for other lymphoid malignancies with plasmacytic features. Notably, multiple myeloma (MM) expresses high levels of ADK, and 6-ETI was toxic to MM cell lines and primary specimens and had a robust antitumor effect in a disseminated MM mouse model. Several nucleoside analogs are effective in treating leukemias and T cell lymphomas, and 6-ETI may fill this niche for the treatment of PEL, plasmablastic lymphoma, MM, and other ADK-expressing cancers.


Asunto(s)
Adenosina Quinasa/metabolismo , Antineoplásicos/farmacología , Linfoma de Efusión Primaria/tratamiento farmacológico , Nucleósidos de Purina/farmacología , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Humanos , Concentración 50 Inhibidora , Linfoma de Efusión Primaria/enzimología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Sci Rep ; 6: 39144, 2016 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-28004818

RESUMEN

Cancer cells have been increasingly grown in pharmaceutical research to understand tumorigenesis and develop new therapeutic drugs. Currently, cells are typically grown using two-dimensional (2-D) cell culture approaches, where the native tumor microenvironment is difficult to recapitulate. Thus, one of the main obstacles in oncology is the lack of proper infection models that recount main features present in tumors. In recent years, microtechnology-based platforms have been employed to generate three-dimensional (3-D) models that better mimic the native microenvironment in cell culture. Here, we present an innovative approach to culture Kaposi's sarcoma-associated herpesvirus (KSHV) infected human B cells in 3-D using a microwell array system. The results demonstrate that the KSHV-infected B cells can be grown up to 15 days in a 3-D culture. Compared with 2-D, cells grown in 3-D had increased numbers of KSHV latency-associated nuclear antigen (LANA) dots, as detected by immunofluorescence microscopy, indicating a higher viral genome copy number. Cells in 3-D also demonstrated a higher rate of lytic reactivation. The 3-D microwell array system has the potential to improve 3-D cell oncology models and allow for better-controlled studies for drug discovery.


Asunto(s)
Antígenos Virales/genética , Linfocitos B/virología , Técnicas de Cultivo de Célula/instrumentación , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/fisiología , Proteínas Nucleares/genética , Antígenos Virales/metabolismo , Línea Celular Tumoral , Humanos , Proteínas Nucleares/metabolismo , Carga Viral , Activación Viral , Latencia del Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...