Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.103
Filtrar
1.
Cell Host Microbe ; 32(5): 755-767.e4, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38653242

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Hemangiossarcoma , Herpesvirus Humano 8 , Camundongos Transgênicos , Sarcoma de Kaposi , Animais , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Camundongos , Hemangiossarcoma/virologia , Hemangiossarcoma/genética , Hemangiossarcoma/patologia , Sarcoma de Kaposi/virologia , Sarcoma de Kaposi/patologia , Genoma Viral , Humanos , Antígenos Virais/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Ganciclovir/uso terapêutico , Ganciclovir/farmacologia , Interleucina-10/genética
2.
BMC Infect Dis ; 24(1): 454, 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38684967

RESUMO

BACKGROUND: Clinically, most patients with Kaposi's sarcoma (KS) are male, and several direct and indirect mechanisms may underlie this increased susceptibility in men, Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is considered to be the primary etiological agent responsible for KS. Thus, we propose the hypothesis that men are more susceptible to HHV-8 infection, leading to a higher incidence of Kaposi's sarcoma among males. A meta-analysis was conducted to evaluate the association between gender and HHV-8 seropositivity in the general population. METHODS: A comprehensive literature search was performed using 6 online databases: PubMed, EMBASE, Cochrane library, Web of Science, CNKI, and Wanfang. Studies published before March 15, 2023, were included. RESULTS: In all, 33 articles including 41 studies were included in the meta-analysis. In the included adult population. men had a higher risk of HHV-8 infection than did women in adult populations from all over the world (odds ratio [OR]: 1.08, 95% confidence interval [CI]: 1.01-1.15), but no differences were found in child population from all over the world (OR: 0.90, 95% CI: 0.79-1.01). There was a significant difference in HHV-8 seroprevalence between men and women in sub-Saharan Africa (SSA) adult population (OR: 1.15, 95% CI: 1.05-1.26). However, no significant differences were observed in sub-Saharan Africa (SSA) child population (OR: 0.90, 95%CI 0.78-1.03). As for other continents, the results showed no significant difference, such as the Asian population (OR: 1.03, 95%CI: 0.92-1.16). or the European and American populations (OR 1.01, 95%CI 0.87-1.17). CONCLUSION: There was a slight gender disparity for HHV-8 infection in the adult population. Among the adult populations from SSA and globally, men were more likely to be infected with HHV-8 than were women. However, no statistical significance was observed in the child populations from SSA and globally. In the future, the inclusion of more standardized studies may strengthen the results of this study.


Assuntos
Infecções por Herpesviridae , Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Masculino , Feminino , Infecções por Herpesviridae/epidemiologia , Infecções por Herpesviridae/virologia , Sarcoma de Kaposi/epidemiologia , Sarcoma de Kaposi/virologia , Fatores Sexuais , Adulto , Incidência , Fatores de Risco , Criança
3.
J Virol ; 98(2): e0126823, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38240588

RESUMO

Protein knockdown with an inducible degradation system is a powerful tool for studying proteins of interest in living cells. Here, we adopted the auxin-inducible degron (AID) approach to detail Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA) function in latency maintenance and inducible viral lytic gene expression. We fused the mini-auxin-inducible degron (mAID) tag at the LANA N-terminus with KSHV bacterial artificial chromosome 16 recombination, and iSLK cells were stably infected with the recombinant KSHV encoding mAID-LANA. Incubation with 5-phenyl-indole-3-acetic acid, a derivative of natural auxin, rapidly degraded LANA within 1.5 h. In contrast to our hypothesis, depletion of LANA alone did not trigger lytic reactivation but rather decreased inducible lytic gene expression when we stimulated reactivation with a combination of ORF50 protein expression and sodium butyrate. Decreased overall lytic gene induction seemed to be associated with a rapid loss of KSHV genomes in the absence of LANA. The rapid loss of viral genomic DNA was blocked by a lysosomal inhibitor, chloroquine. Furthermore, siRNA-mediated knockdown of cellular innate immune proteins, cyclic AMP-GMP synthase (cGAS) and simulator of interferon genes (STING), and other autophagy-related genes rescued the degradation of viral genomic DNA upon LANA depletion. Reduction of the viral genome was not observed in 293FT cells that lack the expression of cGAS. These results suggest that LANA actively prevents viral genomic DNA from sensing by cGAS-STING signaling axis, adding novel insights into the role of LANA in latent genome maintenance.IMPORTANCESensing of pathogens' components is a fundamental cellular immune response. Pathogens have therefore evolved strategies to evade such cellular immune responses. KSHV LANA is a multifunctional protein and plays an essential role in maintaining the latent infection by tethering viral genomic DNA to the host chromosome. We adopted the inducible protein knockdown approach and found that depletion of LANA induced rapid degradation of viral genomic DNA, which is mediated by innate immune DNA sensors and autophagy pathway. These observations suggest that LANA may play a role in hiding KSHV episome from innate immune DNA sensors. Our study thus provides new insights into the role of LANA in latency maintenance.


Assuntos
Antígenos Virais , Herpesvirus Humano 8 , Plasmídeos , Sarcoma de Kaposi , Humanos , Antígenos Virais/metabolismo , DNA , Herpesvirus Humano 8/fisiologia , Ácidos Indolacéticos , Nucleotidiltransferases/genética , Sarcoma de Kaposi/virologia , Latência Viral , Proteínas Nucleares/metabolismo
4.
J Virol ; 97(10): e0083223, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37796128

RESUMO

IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of several B cell malignancies and Kaposi's sarcoma. We analyzed the function of K8.1, the major antigenic component of the KSHV virion in the infection of different cells. To do this, we deleted K8.1 from the viral genome. It was found that K8.1 is critical for the infection of certain epithelial cells, e.g., a skin model cell line but not for infection of many other cells. K8.1 was found to mediate attachment of the virus to cells where it plays a role in infection. In contrast, we did not find K8.1 or a related protein from a closely related monkey virus to activate fusion of the viral and cellular membranes, at least not under the conditions tested. These findings suggest that K8.1 functions in a highly cell-specific manner during KSHV entry, playing a crucial role in the attachment of KSHV to, e.g., skin epithelial cells.


Assuntos
Glicoproteínas , Herpesvirus Humano 8 , Queratinócitos , Proteínas Virais , Ligação Viral , Internalização do Vírus , Humanos , Glicoproteínas/deficiência , Glicoproteínas/genética , Glicoproteínas/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Queratinócitos/metabolismo , Queratinócitos/virologia , Sarcoma de Kaposi/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Fusão de Membrana , Pele/citologia
5.
J Virol ; 97(10): e0063723, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37750723

RESUMO

IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is a human herpesvirus associated with several human cancers, typically in patients with compromised immune systems. Herpesviruses establish lifelong infections in hosts in part due to the two phases of infection: the dormant and active phases. Effective antiviral treatments to prevent the production of new viruses are needed to treat KSHV. A detailed microscopy-based investigation of the molecular interactions between viral protein and viral DNA revealed how protein-protein interactions play a role in DNA-binding specificity. This analysis will lead to a more in-depth understanding of KSHV DNA replication and serve as the basis for anti-viral therapies that disrupt and prevent the protein-DNA interactions, thereby decreasing spread to new hosts.


Assuntos
DNA Viral , Herpesvirus Humano 8 , Microscopia Eletrônica , Multimerização Proteica , Transativadores , Humanos , Sítios de Ligação , DNA Viral/química , DNA Viral/metabolismo , DNA Viral/ultraestrutura , Herpesvirus Humano 8/química , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/ultraestrutura , Ligação Proteica , Mapas de Interação de Proteínas , Especificidade por Substrato , Transativadores/química , Transativadores/metabolismo , Transativadores/ultraestrutura , Replicação Viral/genética , Sarcoma de Kaposi/virologia
7.
J Virol ; 97(3): e0176322, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36995092

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi sarcoma (KS), the plasmablastic form of multicentric Castleman's disease, and primary effusion lymphoma. In sub-Saharan Africa, KS is the most common HIV-related malignancy and one of the most common childhood cancers. Immunosuppressed patients, including HIV-infected patients, are more prone to KSHV-associated disease. KSHV encodes a viral protein kinase (vPK) that is expressed from ORF36. KSHV vPK contributes to the optimal production of infectious viral progeny and upregulation of protein synthesis. To elucidate the interactions of vPK with cellular proteins in KSHV-infected cells, we used a bottom-up proteomics approach and identified host protein ubiquitin-specific peptidase 9X-linked (USP9X) as a potential interactor of vPK. Subsequently, we validated this interaction using a co-immunoprecipitation assay. We report that both the ubiquitin-like and the catalytic domains of USP9X are important for association with vPK. To uncover the biological relevance of the USP9X/vPK interaction, we investigated whether the knockdown of USP9X would modulate viral reactivation. Our data suggest that depletion of USP9X inhibits both viral reactivation and the production of infectious virions. Understanding how USP9X influences the reactivation of KSHV will provide insights into how cellular deubiquitinases regulate viral kinase activity and how viruses co-opt cellular deubiquitinases to propagate infection. Hence, characterizing the roles of USP9X and vPK during KSHV infection constitutes a first step toward identifying a potentially critical interaction that could be targeted by future therapeutics. IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi sarcoma (KS), the plasmablastic form of multicentric Castleman's disease, and primary effusion lymphoma. In sub-Saharan Africa, KS is the most common HIV-related malignancy. KSHV encodes a viral protein kinase (vPK) that aids viral replication. To elucidate the interactions of vPK with cellular proteins in KSHV-infected cells, we used an affinity purification approach and identified host protein ubiquitin-specific peptidase 9X-linked (USP9X) as a potential interactor of vPK. Depletion of USP9X inhibits both viral reactivation and the production of infectious virions. Overall, our data suggest a proviral role for USP9X.


Assuntos
Herpesvirus Humano 8 , Sarcoma de Kaposi , Ubiquitina Tiolesterase , Criança , Humanos , Enzimas Desubiquitinantes , Herpesvirus Humano 8/fisiologia , Infecções por HIV/complicações , Linfoma de Efusão Primária , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Ubiquitina Tiolesterase/genética , Proteínas Virais/genética
8.
Rev. peru. med. exp. salud publica ; 39(3): 352-356, jul.-sep. 2022. tab
Artigo em Espanhol | LILACS | ID: biblio-1410012

RESUMO

RESUMEN El sarcoma de Kaposi (SK) es el cáncer más frecuente en las personas que viven con VIH. Las investigaciones sobre esta condición son escasas en la región, por lo que, el objetivo de este artículo fue describir las características demográficas, clínicas y terapéuticas de los pacientes con VIH que desarrollaron SK en el Hospital Cayetano Heredia entre el 2000 y 2018. Se identificaron 129 casos de SK, con una mediana de edad de 33 años, con predominio en varones con el 92% (119/129), y en su mayoría hombres que tienen sexo con hombres (HSH). La mediana de tiempo desde el diagnóstico de VIH hasta el del SK fue de cinco meses, asociado con un recuento de linfocitos CD4 de 64 células/µL (RIC: 33-185) al momento del diagnóstico de SK. El compromiso cutáneo fue el más común; sin embargo, al menos la mitad de ellos también tuvo la forma visceral.


ABSTRACT Kaposi's sarcoma (KS) is the most frequent cancer in people living with HIV. Research on this condition is scarce in the region, therefore, this article aimed to describe the demographic, clinical and therapeutic characteristics of patients with HIV who developed KS at the Cayetano Heredia Hospital between 2000 and 2018. A total of 129 KS cases were identified, with a median age of 33 years, predominantly males with 92% (119/129), and mostly men who have sex with men (MSM). The median time from HIV diagnosis to KS diagnosis was five months, associated with a CD4 lymphocyte count of 64 cells/μL (IQR: 33-185) at KS diagnosis. Cutaneous involvement was the most common presentation; however, at least half also had the visceral form.


Assuntos
Humanos , Masculino , Feminino , Adulto , Sarcoma de Kaposi/epidemiologia , Infecções Oportunistas Relacionadas com a AIDS/epidemiologia , Peru/epidemiologia , Sarcoma de Kaposi/virologia , Estudos de Coortes , Infecções Oportunistas Relacionadas com a AIDS/virologia , Contagem de Linfócito CD4 , Carga Viral , Distribuição por Idade e Sexo
9.
J Biol Chem ; 298(6): 102012, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35525271

RESUMO

Constitutive activation of the canonical NF-κB signaling pathway is a major factor in Kaposi's sarcoma-associated herpes virus pathogenesis where it is essential for the survival of primary effusion lymphoma. Central to this process is persistent upregulation of the inhibitor of κB kinase (IKK) complex by the virally encoded oncoprotein vFLIP. Although the physical interaction between vFLIP and the IKK kinase regulatory component essential for persistent activation, IKKγ, has been well characterized, it remains unclear how the kinase subunits are rendered active mechanistically. Using a combination of cell-based assays, biophysical techniques, and structural biology, we demonstrate here that vFLIP alone is sufficient to activate the IKK kinase complex. Furthermore, we identify weakly stabilized, high molecular weight vFLIP-IKKγ assemblies that are key to the activation process. Taken together, our results are the first to reveal that vFLIP-induced NF-κB activation pivots on the formation of structurally specific vFLIP-IKKγ multimers which have an important role in rendering the kinase subunits active through a process of autophosphorylation. This mechanism of NF-κB activation is in contrast to those utilized by endogenous cytokines and cellular FLIP homologues.


Assuntos
Herpesvirus Humano 8 , Sarcoma de Kaposi , Ativação Enzimática/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Quinase I-kappa B/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas Oncogênicas/metabolismo , Sarcoma de Kaposi/enzimologia , Sarcoma de Kaposi/virologia , Proteínas Virais/metabolismo
10.
Pathol Oncol Res ; 28: 1610055, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35140551

RESUMO

MicroRNAs (miRNAs) are the non-coding RNAs that can both attach to the untranslated and coding sections of target mRNAs, triggering destruction or post-transcriptional alteration. miRNAs regulate various cellular processes such as immune function, apoptosis, and tumorigenesis. About 35,000 miRNAs have been discovered in the human genome. The increasing evidence suggests that the dysregulation of human miRNAs may have a role in the etiology of some disorders including cancer. Only a small sub-set of human miRNAs has functionally been validated in the pathogenesis of oncogenic viruses such as Kaposi's sarcoma-associated herpesvirus (KSHV). KSHV is the cause of various human malignancies including primary effusion lymphoma (PEL) and Kaposi's sarcoma (KS), which are mainly seen in AIDS patients or other immunocompromised people. We aimed to identify the miRNAs in Kaposi's sarcoma cases, with the comparison of KSHV seropositive and seronegative tumors with the controls and in each other in Turkish Kaposi's sarcoma patients. We performed the miRNA-sequencing at genome level in the peripheral blood mononuclear cells of 16 Kaposi's sarcoma patients, and in 8 healthy controls matched for age, gender, and ethnicity. A total of 642 miRNA molecules with different expression profiles were identified between the patients and the healthy controls. Currently, out of 642 miRNAs, 7 miRNAs (miR-92b-3p, miR-490-3p, miR-615-3p, miR-629-5p, miR-1908, miR-3180, miR-4433b-3p) which have not been described in the literature in the context of Kaposi's sarcoma were addressed in the study for the first time and 9 novel miRNAs, not found previously in the database, have been detected in Kaposi's sarcoma using the miRNA-sequencing technique. This study demonstrates the identification of differently expressed miRNAs which might be the new therapeutic targets for Kaposi's sarcoma, that has limited treatment options and can be used in the etiology, diagnosis, and prognosis of this cancer.


Assuntos
MicroRNAs/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virologia , Herpesvirus Humano 8 , Humanos , MicroRNAs/análise , Análise de Sequência de RNA
11.
Virology ; 568: 101-114, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35152042

RESUMO

Kaposi sarcoma (KS)-associated herpesvirus (KSHV/HHV-8) was first sequenced from the body cavity (BC) lymphoma cell line, BC-1, in 1996. Few other KSHV genomes have been reported. Our knowledge of sequence variation for this virus remains spotty. This study reports additional genomes from historical US patient samples and from African KS biopsies. It describes an assay that spans regions of the virus that cannot be covered by short read sequencing. These include the terminal repeats, the LANA repeats, and the origins of replication. A phylogenetic analysis, based on 107 genomes, identified three distinct clades; one containing isolates from USA/Europe/Japan collected in the 1990s and two of Sub-Saharan Africa isolates collected since 2010. This analysis indicates that the KSHV strains circulating today differ from the isolates collected at the height of the AIDS epidemic. This analysis helps experimental designs and potential vaccine studies.


Assuntos
Genoma Viral , Genômica , Genótipo , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/classificação , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/virologia , Adulto , Linhagem Celular , Feminino , Regulação Viral da Expressão Gênica , Genômica/métodos , Infecções por Herpesviridae/diagnóstico , Herpesvirus Humano 8/isolamento & purificação , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Filogenia , Recombinação Genética
12.
Nat Commun ; 13(1): 472, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35078976

RESUMO

The Kaposi's sarcoma associated herpesvirus protein ORF45 binds the extracellular signal-regulated kinase (ERK) and the p90 Ribosomal S6 kinase (RSK). ORF45 was shown to be a kinase activator in cells but a kinase inhibitor in vitro, and its effects on the ERK-RSK complex are unknown. Here, we demonstrate that ORF45 binds ERK and RSK using optimized linear binding motifs. The crystal structure of the ORF45-ERK2 complex shows how kinase docking motifs recognize the activated form of ERK. The crystal structure of the ORF45-RSK2 complex reveals an AGC kinase docking system, for which we provide evidence that it is functional in the host. We find that ORF45 manipulates ERK-RSK signaling by favoring the formation of a complex, in which activated kinases are better protected from phosphatases and docking motif-independent RSK substrate phosphorylation is selectively up-regulated. As such, our data suggest that ORF45 interferes with the natural design of kinase docking systems in the host.


Assuntos
Cristalografia por Raios X/métodos , Herpesvirus Humano 8/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/química , Proteínas Quinases S6 Ribossômicas 90-kDa/química , Sarcoma de Kaposi/metabolismo , Linhagem Celular , Biologia Computacional , Herpesvirus Humano 8/química , Herpesvirus Humano 8/isolamento & purificação , Humanos , Proteínas Imediatamente Precoces/química , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosforilação , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Transdução de Sinais
13.
J Virol ; 96(5): e0156021, 2022 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-34936820

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is the cause of several human cancers, including the endothelial cell (EC) malignancy, Kaposi's sarcoma. Unique KSHV genes absent from other human herpesvirus genomes, the "K-genes," are important for KSHV replication and pathogenesis. Among these, the kaposin transcript is highly expressed in all phases of infection, but its complex polycistronic nature has hindered functional analysis to date. At least three proteins are produced from the kaposin transcript: Kaposin A (KapA), B (KapB), and C (KapC). To determine the relative contributions of kaposin proteins during KSHV infection, we created a collection of mutant viruses unable to produce kaposin proteins individually or in combination. In previous work, we showed KapB alone recapitulated the elevated proinflammatory cytokine transcripts associated with KS via the disassembly of RNA granules called processing bodies (PBs). Using the new ΔKapB virus, we showed that KapB was necessary for this effect during latent KSHV infection. Moreover, we observed that despite the ability of all kaposin-deficient latent iSLK cell lines to produce virions, all displayed low viral episome copy number, a defect that became more pronounced after primary infection of naive ECs. For ΔKapB, provision of KapB in trans failed to complement the defect, suggesting a requirement for the kaposin locus in cis. These findings demonstrate that our panel of kaposin-deficient viruses enables precise analysis of the respective contributions of individual kaposin proteins to KSHV replication. Moreover, our mutagenesis approach serves as a guide for the functional analysis of other complex multicistronic viral loci. IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) expresses high levels of the kaposin transcript during both latent and lytic phases of replication. Due to its repetitive, GC-rich nature and polycistronic coding capacity, until now no reagents existed to permit a methodical analysis of the role of individual kaposin proteins in KSHV replication. We report the creation of a panel of recombinant viruses and matched producer cell lines that delete kaposin proteins individually or in combination. We demonstrate the utility of this panel by confirming the requirement of one kaposin translation product to a key KSHV latency phenotype. This study describes a new panel of molecular tools for the KSHV field to enable precise analysis of the roles of individual kaposin proteins during KSHV infection.


Assuntos
Herpesvirus Humano 8 , Sarcoma de Kaposi , Proteínas Virais , Linhagem Celular Tumoral , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Mutação , Sarcoma de Kaposi/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Latência Viral/genética
14.
J Clin Oncol ; 40(3): 294-306, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-34890242

RESUMO

The observation in 1981 of the emergence of Kaposi sarcoma (KS) among young men who had sex with men was one of the first harbingers of the HIV epidemic. With advances in HIV care, the incidence of HIV-associated KS (HIV+KS) has decreased over time in the United States. However, it remains a persistent malignancy among some HIV-infected populations and is one of the most common tumors in sub-Saharan Africa. Because of the relapsing and remitting nature of this cancer, patients with HIV+KS can experience significant, long-term, morbidity. Patients with severe HIV+KS may also have concurrent lymphoproliferative syndromes, malignancies, and/or infections that can contribute to mortality. Several chemotherapy agents were explored in clinical trials for HIV+KS during the early stage of the epidemic. As HIV+KS emerges with CD4 lymphopenia and immunodysregulation, T-cell-sparing options are important to consider. Here, we explore the pathogenesis of HIV+KS and the current evidence for immunotherapy and therapies that potentially target KS pathogenesis. This review provides the current landscape of therapies for HIV+KS and highlights management issues for patients with HIV and cancer.


Assuntos
Antineoplásicos/uso terapêutico , Infecções por HIV , Imunoterapia , Oncologia/tendências , Sarcoma de Kaposi/terapia , Antineoplásicos/efeitos adversos , Difusão de Inovações , Infecções por HIV/epidemiologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Imunoterapia/efeitos adversos , Terapia de Alvo Molecular , Sarcoma de Kaposi/epidemiologia , Sarcoma de Kaposi/imunologia , Sarcoma de Kaposi/virologia , Resultado do Tratamento
15.
Blood ; 139(7): 1013-1025, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34479367

RESUMO

Kaposi sarcoma (KS) herpesvirus (KSHV), also known as human herpesvirus 8, is the causal agent of KS but is also pathogenetically related to several lymphoproliferative disorders, including primary effusion lymphoma (PEL)/extracavitary (EC) PEL, KSHV-associated multicentric Castleman disease (MCD), KSHV+ diffuse large B-cell lymphoma, and germinotropic lymphoproliferative disorder. These different KSHV-associated diseases may co-occur and may have overlapping features. KSHV, similar to Epstein-Barr virus (EBV), is a lymphotropic gammaherpesvirus that is preferentially present in abnormal lymphoid proliferations occurring in immunecompromised individuals. Notably, both KSHV and EBV can infect and transform the same B cell, which is frequently seen in KSHV+ EBV+ PEL/EC-PEL. The mechanisms by which KSHV leads to lymphoproliferative disorders is thought to be related to the expression of a few transforming viral genes that can affect cellular proliferation and survival. There are critical differences between KSHV-MCD and PEL/EC-PEL, the 2 most common KSHV-associated lymphoid proliferations, including viral associations, patterns of viral gene expression, and cellular differentiation stage reflected by the phenotype and genotype of the infected abnormal B cells. Advances in treatment have improved outcomes, but mortality rates remain high. Our deepening understanding of KSHV biology, clinical features of KSHV-associated diseases, and newer clinical interventions should lead to improved and increasingly targeted therapeutic interventions.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Doenças Hematológicas/patologia , Herpesvirus Humano 4/isolamento & purificação , Herpesvirus Humano 8/isolamento & purificação , Transtornos Linfoproliferativos/patologia , Sarcoma de Kaposi/complicações , Infecções por Vírus Epstein-Barr/virologia , Doenças Hematológicas/epidemiologia , Doenças Hematológicas/virologia , Humanos , Transtornos Linfoproliferativos/epidemiologia , Transtornos Linfoproliferativos/virologia , Sarcoma de Kaposi/virologia
16.
Medicine (Baltimore) ; 100(51): e28328, 2021 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-34941134

RESUMO

ABSTRACT: Pulmonary Kaposi sarcoma (pKS) caused by Human herpesvirus 8 (HHV-8) is a devastating form of KS in patients with advanced acquired immunodeficiency syndrome (AIDS) and is associated with increased morbidity and mortality. Blood T cells play a central role in the response of HIV-1 and HHV-8. However, little information is available on T cells in the alveolar space of HIV-1-associated pKS patients.Therefore, we examined CD8+ and CD4+ T cells in the alveolar space in comparison with the blood of patients with pKS. We recruited 26 HIV-1 positive patients with KS, including 15 patients with pKS. Bronchoalveolar lavage (BAL) cells and blood mononuclear cells were analyzed for T cell memory phenotypes, surface markers associated with exhaustion, and intracellular cytokine staining (ICS) using flow cytometry. HIV-1 and HHV-8 viral loads were measured in plasma by quantitative PCR.BAL T cells showed reduced inflammatory capacities and significantly diminished polyfunctionality compared to blood T cells from patients with pKS. This was not accompanied by increased expression of exhaustion markers, such as TIM-3 and PD-1.More importantly, we found a negative correlation between the production of MIP1-ß and TNF-α in T cells in BAL and blood, indicating compartmentalised immune responses to pKS and accentuated chronic HIV-1/HHV-8 pathogenesis via T cells in the lungs of people with pKS.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/virologia , Líquido da Lavagem Broncoalveolar/virologia , Soropositividade para HIV/complicações , Herpesvirus Humano 8/imunologia , Neoplasias Pulmonares/virologia , Sarcoma de Kaposi/virologia , Linfócitos T Reguladores/imunologia , Antígenos Virais/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , HIV-1/patogenicidade , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/isolamento & purificação , Humanos , Reação em Cadeia da Polimerase
17.
Cell Rep ; 37(13): 110144, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34965440

RESUMO

Kaposi's sarcoma herpesvirus (KSHV) is an angiogenesis-inducing oncovirus whose ability to usurp the oxygen-sensing machinery is central to its oncogenicity. By upregulating the hypoxia-inducible factors (HIFs), KSHV reprograms infected cells to a hypoxia-like state, triggering angiogenesis. Here we identify a link between KSHV replicative biology and oncogenicity by showing that KSHV's ability to regulate HIF2α levels and localization to the endoplasmic reticulum (ER) in normoxia enables translation of viral lytic mRNAs through the HIF2α-regulated eIF4E2 translation-initiation complex. This mechanism of translation in infected cells is critical for lytic protein synthesis and contributes to KSHV-induced PDGFRA activation and VEGF secretion. Thus, KSHV regulation of the oxygen-sensing machinery allows virally infected cells to initiate translation via the mTOR-dependent eIF4E1 or the HIF2α-dependent, mTOR-independent, eIF4E2. This "translation initiation plasticity" (TRIP) is an oncoviral strategy used to optimize viral protein expression that links molecular strategies of viral replication to angiogenicity and oncogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinogênese/patologia , Herpesvirus Humano 8/fisiologia , Hipóxia/fisiopatologia , Iniciação Traducional da Cadeia Peptídica , Sarcoma de Kaposi/patologia , Replicação Viral , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Carcinogênese/genética , Carcinogênese/metabolismo , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Humanos , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Ativação Viral
18.
mBio ; 12(6): e0343121, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34933446

RESUMO

Infection by Kaposi's sarcoma-associated herpesvirus (KSHV) is causally associated with numerous cancers. The mechanism of KSHV-induced oncogenesis remains unclear. By performing a CRISPR-Cas9 screening in a model of KSHV-induced cellular transformation of primary cells, we identified epigenetic regulators that were essential for KSHV-induced cellular transformation. Examination of TCGA data sets of the top 9 genes, including glutamate-rich WD repeat containing 1 (GRWD1), a WD40 family protein upregulated by KSHV, that had positive effects on cell proliferation and survival of KSHV-transformed cells (KMM) but not the matched primary cells (MM), uncovered the predictive values of their expressions for patient survival in numerous types of cancer. We revealed global epigenetic remodeling including H3K4me3 epigenetic active mark in KMM cells compared to MM cells. Knockdown of GRWD1 inhibited cell proliferation, cellular transformation, and tumor formation and caused downregulation of global H3K4me3 mark in KMM cells. GRWD1 interacted with WD repeat domain 5 (WDR5), the core protein of H3K4 methyltransferase complex, and several H3K4me3 methyltransferases, including myeloid leukemia 2 (MLL2). Knockdown of WDR5 and MLL2 phenocopied GRWD1 knockdown, caused global reduction of H3K4me3 mark, and altered the expression of similar sets of genes. Transcriptome sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) analyses further identified common and distinct cellular genes and pathways that were regulated by GRWD1, WDR5, and MLL2. These results indicate that KSHV hijacks the GRWD1-WDR5-MLL2 epigenetic complex to regulate H3K4me3 methylation of specific genes, which is essential for KSHV-induced cellular transformation. Our work has identified an epigenetic complex as a novel therapeutic target for KSHV-induced cancers. IMPORTANCE By performing a genome-wide CRISPR-Cas9 screening, we have identified cellular epigenetic regulators that are essential for KSHV-induced cellular transformation. Among them, GRWD1 regulates epigenetic active mark H3K4me3 by interacting with WDR5 and MLL2 and recruiting them to chromatin loci of specific genes in KSHV-transformed cells. Hence, KSHV hijacks the GRWD1-WDR5-MLL2 complex to remodel cellular epigenome and induce cellular transformation. Since the dysregulation of GRWD1 is associated with poor prognosis in several types of cancer, GRWD1 might also be a critical driver in other viral or nonviral cancers.


Assuntos
Proteínas de Transporte/metabolismo , Transformação Celular Viral , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Herpesvirus Humano 8/fisiologia , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Sarcoma de Kaposi/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Ligação a DNA/genética , Herpesvirus Humano 8/genética , Histonas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Ligação Proteica , Sarcoma de Kaposi/enzimologia , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virologia
19.
PLoS Pathog ; 17(12): e1009600, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34936683

RESUMO

Kaposi's sarcoma (KS) is an angioproliferative and invasive tumor caused by Kaposi's sarcoma-associated herpesvirus (KSHV). The cellular origin of KS tumor cells remains contentious. Recently, evidence has accrued indicating that KS may arise from KSHV-infected mesenchymal stem cells (MSCs) through mesenchymal-to-endothelial transition (MEndT), but the transformation process has been largely unknown. In this study, we investigated the KSHV-mediated MEndT process and found that KSHV infection rendered MSCs incomplete endothelial lineage differentiation and formed hybrid mesenchymal/endothelial (M/E) state cells characterized by simultaneous expression of mesenchymal markers Nestin/PDGFRA/α-SAM and endothelial markers CD31/PDPN/VEGFR2. The hybrid M/E cells have acquired tumorigenic phenotypes in vitro and the potential to form KS-like lesions after being transplanted in mice under renal capsules. These results suggest a homology of KSHV-infected MSCs with Kaposi's sarcoma where proliferating KS spindle-shaped cells and the cells that line KS-specific aberrant vessels were also found to exhibit the hybrid M/E state. Furthermore, the genetic analysis identified KSHV-encoded FLICE inhibitory protein (vFLIP) as a crucial regulator controlling KSHV-induced MEndT and generating hybrid M/E state cells for tumorigenesis. Overall, KSHV-mediated MEndT that transforms MSCs to tumorigenic hybrid M/E state cells driven by vFLIP is an essential event in Kaposi's sarcomagenesis.


Assuntos
Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Nestina/metabolismo , Sarcoma de Kaposi/virologia , Proteínas Virais/metabolismo , Animais , Carcinogênese , Diferenciação Celular , Células Endoteliais/patologia , Células Endoteliais/virologia , Feminino , Infecções por Herpesviridae/patologia , Herpesvirus Humano 8/fisiologia , Humanos , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/virologia , Camundongos , Nestina/genética , Sarcoma de Kaposi/patologia , Proteínas Virais/genética
20.
Nucleic Acids Res ; 49(22): 13179-13193, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34871450

RESUMO

Cellular and virus-coded long non-coding (lnc) RNAs support multiple roles related to biological and pathological processes. Several lncRNAs sequester their 3' termini to evade cellular degradation machinery, thereby supporting disease progression. An intramolecular triplex involving the lncRNA 3' terminus, the element for nuclear expression (ENE), stabilizes RNA transcripts and promotes persistent function. Therefore, such ENE triplexes, as presented here in Kaposi's sarcoma-associated herpesvirus (KSHV) polyadenylated nuclear (PAN) lncRNA, represent targets for therapeutic development. Towards identifying novel ligands targeting the PAN ENE triplex, we screened a library of immobilized small molecules and identified several triplex-binding chemotypes, the tightest of which exhibits micromolar binding affinity. Combined biophysical, biochemical, and computational strategies localized ligand binding to a platform created near a dinucleotide bulge at the base of the triplex. Crystal structures of apo (3.3 Å) and ligand-soaked (2.5 Å) ENE triplexes, which include a stabilizing basal duplex, indicate significant local structural rearrangements within this dinucleotide bulge. MD simulations and a modified nucleoside analog interference technique corroborate the role of the bulge and the base of the triplex in ligand binding. Together with recently discovered small molecules that reduce nuclear MALAT1 lncRNA levels by engaging its ENE triplex, our data supports the potential of targeting RNA triplexes with small molecules.


Assuntos
Herpesvirus Humano 8/metabolismo , Nucleotídeos/metabolismo , Poli A/metabolismo , RNA Longo não Codificante/metabolismo , RNA Viral/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Sequência de Bases , Cristalografia por Raios X , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Humanos , Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Estrutura Molecular , Conformação de Ácido Nucleico , Nucleotídeos/genética , Poli A/química , Poli A/genética , Estabilidade de RNA/genética , RNA Longo não Codificante/química , RNA Longo não Codificante/genética , RNA Viral/química , RNA Viral/genética , Sarcoma de Kaposi/virologia , Bibliotecas de Moléculas Pequenas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...