Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 174
Filtrar
1.
Cell ; 185(20): 3652-3670, 2022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-36113467

RESUMO

Epstein-Barr virus (EBV) is a ubiquitous, oncogenic virus that is associated with a number of different human malignancies as well as autoimmune disorders. The expression of EBV viral proteins and non-coding RNAs contribute to EBV-mediated disease pathologies. The virus establishes life-long latency in the human host and is adept at evading host innate and adaptive immune responses. In this review, we discuss the life cycle of EBV, the various functions of EBV-encoded proteins and RNAs, the ability of the virus to activate and evade immune responses, as well as the neoplastic and autoimmune diseases that are associated with EBV infection in the human population.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Biologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , RNA/metabolismo , Proteínas Virais/metabolismo , Latência Viral
2.
J Virol ; 98(1): e0179123, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38168672

RESUMO

In the United States (US), biosafety and biosecurity oversight of research on viruses is being reappraised. Safety in virology research is paramount and oversight frameworks should be reviewed periodically. Changes should be made with care, however, to avoid impeding science that is essential for rapidly reducing and responding to pandemic threats as well as addressing more common challenges caused by infectious diseases. Decades of research uniquely positioned the US to be able to respond to the COVID-19 crisis with astounding speed, delivering life-saving vaccines within a year of identifying the virus. We should embolden and empower this strength, which is a vital part of protecting the health, economy, and security of US citizens. Herein, we offer our perspectives on priorities for revised rules governing virology research in the US.


Assuntos
Pesquisa Biomédica , Contenção de Riscos Biológicos , Virologia , Humanos , COVID-19 , Estados Unidos , Vírus , Pesquisa Biomédica/normas
3.
Proc Natl Acad Sci U S A ; 119(29): e2200206119, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858339

RESUMO

Human papillomaviruses (HPVs) infect the basal proliferating cells of the stratified epithelium, but the productive phase of the life cycle (consisting of viral genome amplification, late gene expression, and virion assembly) is restricted to the highly differentiated suprabasal cells. While much is known regarding the mechanisms that HPVs use to block activation of an innate immune response in undifferentiated cells, little is known concerning how HPV prevents an interferon (IFN) response upon differentiation. Here, we demonstrate that high-risk HPVs hijack a natural function of apoptotic caspases to suppress an IFN response in differentiating epithelial cells. We show that caspase inhibition results in the secretion of type I and type III IFNs that can act in a paracrine manner to induce expression of interferon-stimulated genes (ISGs) and block productive replication of HPV31. Importantly, we demonstrate that the expression of IFNs is triggered by the melanoma differentiation-associated gene 5 (MDA5)-mitochondrial antiviral-signaling protein (MAVS)-TBK1 (TANK-binding kinase 1) pathway, signifying a response to double-stranded RNA (dsRNA). Additionally, we identify a role for MDA5 and MAVS in restricting productive viral replication during the normal HPV life cycle. This study identifies a mechanism by which HPV reprograms the cellular environment of differentiating cells through caspase activation, co-opting a nondeath function of proteins normally involved in apoptosis to block antiviral signaling and promote viral replication.


Assuntos
Caspases , Papillomavirus Humano 31 , Helicase IFIH1 Induzida por Interferon , Interferons , Infecções por Papillomavirus , Replicação Viral , Caspases/metabolismo , Papillomavirus Humano 31/fisiologia , Humanos , Helicase IFIH1 Induzida por Interferon/metabolismo , Interferons/metabolismo , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/virologia
4.
Br J Haematol ; 205(1): 194-206, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38769021

RESUMO

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of lymphoma worldwide, accounting for up to 40% of new non-Hodgkin Lymphoma (NHL) globally. People living with HIV are up to 17 times more likely to develop NHL, and as such, DLBCL is the leading cause of cancer death in this high-risk population. While histologically indistinguishable, HIV-associated (HIV+) and HIV-negative (HIV-) DLBCL are molecularly distinct, and biological differences may have implications for the development of future therapeutic interventions. Further, the impact of immunologic differences in people with HIV, including preceding ART, remains largely unknown. Here, we investigate the impact of HIV infection and ART exposure on the clinical features of DLBCL and T-cell immune response by performing imaging mass cytometry on our unique patient cohort in Malawi. In this cohort, HIV infection is positively prognostic, and HIV+/ART-naïve patients have the best outcomes. No established biomarkers other than Ki67 are associated with HIV or ART status, and the only tumour-intrinsic biomarkers that remain prognostic are MYC and MYC/BCL2 protein co-expression. Finally, TCR clonality is associated with distinct tumour-T cell interactions by HIV/ART status, indicating differential anti-tumour immune responses. We demonstrate previously undescribed HIV and ART-related differences in the DLBCL tumour microenvironment.


Assuntos
Infecções por HIV , Linfoma Difuso de Grandes Células B , Humanos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Linfócitos T/imunologia , Antirretrovirais/uso terapêutico
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
6.
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
7.
J Virol ; 97(2): e0008923, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36700640

RESUMO

Viruses have brought humanity many challenges: respiratory infection, cancer, neurological impairment and immunosuppression to name a few. Virology research over the last 60+ years has responded to reduce this disease burden with vaccines and antivirals. Despite this long history, the COVID-19 pandemic has brought unprecedented attention to the field of virology. Some of this attention is focused on concern about the safe conduct of research with human pathogens. A small but vocal group of individuals has seized upon these concerns - conflating legitimate questions about safely conducting virus-related research with uncertainties over the origins of SARS-CoV-2. The result has fueled public confusion and, in many instances, ill-informed condemnation of virology. With this article, we seek to promote a return to rational discourse. We explain the use of gain-of-function approaches in science, discuss the possible origins of SARS-CoV-2 and outline current regulatory structures that provide oversight for virological research in the United States. By offering our expertise, we - a broad group of working virologists - seek to aid policy makers in navigating these controversial issues. Balanced, evidence-based discourse is essential to addressing public concern while maintaining and expanding much-needed research in virology.


Assuntos
Pesquisa , Virologia , Viroses , Humanos , COVID-19/prevenção & controle , Disseminação de Informação , Pandemias/prevenção & controle , Formulação de Políticas , Pesquisa/normas , Pesquisa/tendências , SARS-CoV-2 , Virologia/normas , Virologia/tendências , Viroses/prevenção & controle , Viroses/virologia , Vírus
8.
PLoS Pathog ; 18(11): e1010990, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36417478

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr (EBV) are gammaherpesviruses associated with multiple human malignancies. KSHV is the etiological agent of Kaposi's Sarcoma, primary effusion lymphoma (PEL) and multicentric Castleman's disease (MCD). EBV is associated with Burkitt's lymphoma (BL), Hodgkin's lymphoma (HL), nasopharyngeal carcinoma (NPC) and gastric carcinoma (GC). KSHV and EBV establish life-long latency in the human host with intermittent periods of lytic reactivation. Here, we identified a cellular factor named transforming growth factor-beta regulator 4 (TBRG4) that plays a role in the gammaherpesvirus lifecycle. We find that TBRG4, a protein that is localized to the mitochondria, can regulate lytic reactivation from latency of both KSHV and EBV. Knockdown of TBRG4 in cells latently infected with KSHV or EBV induced viral lytic gene transcription and replication. TBRG4 deficiency causes mitochondrial stress and increases reactive oxygen species (ROS) production. Treatment with a ROS scavenger decreased viral reactivation from latency in TBRG4-depleted cells. These data suggest that TBRG4 serves as a cellular repressor of KSHV and EBV reactivation through the regulation of ROS production.


Assuntos
Herpesvirus Humano 4 , Herpesvirus Humano 8 , Proteínas Mitocondriais , Latência Viral , Humanos , Herpesvirus Humano 4/fisiologia , Herpesvirus Humano 8/fisiologia , Proteínas Mitocondriais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas de Ligação a RNA/metabolismo
9.
Cell Mol Life Sci ; 80(6): 149, 2023 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-37183204

RESUMO

STING acts as a cytosolic nucleotide sensor to trigger host defense upon viral or bacterial infection. While STING hyperactivation can exert anti-tumor effects by increasing T cell filtrates, in other contexts hyperactivation of STING can contribute to autoimmune and neuroinflammatory diseases. Several STING targeting agonists and a smaller subset of antagonists have been developed, yet STING targeted degraders, or PROTACs, remain largely underexplored. Here, we report a series of STING-agonist derived PROTACs that promote STING degradation in renal cell carcinoma (RCC) cells. We show that our STING PROTACs activate STING and target activated/phospho-STING for degradation. Locking STING on the endoplasmic reticulum via site-directed mutagenesis disables STING translocation to the proteasome and resultingly blocks STING degradation. We also demonstrate that PROTAC treatment blocks downstream innate immune signaling events and attenuates the anti-viral response. Interestingly, we find that VHL acts as a bona fide E3 ligase for STING in RCC; thus, VHL-recruiting STING PROTACs further promote VHL-dependent STING degradation. Our study reveals the design and biological assessment of VHL-recruiting agonist-derived STING PROTACs, as well as demonstrates an example of hijacking a physiological E3 ligase to enhance target protein degradation via distinct mechanisms.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Quimera de Direcionamento de Proteólise , Carcinoma de Células Renais/tratamento farmacológico , Ubiquitina-Proteína Ligases/metabolismo , Proteólise , Neoplasias Renais/tratamento farmacológico , Imunidade Inata , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
10.
Int J Cancer ; 153(12): 2082-2092, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-37602960

RESUMO

Kaposi sarcoma (KS) is the most common cancer in people living with HIV (PLWH) in many countries where KS-associated herpesvirus is endemic. Treatment has changed little in 20 years, but the disease presentation has. This prospective cohort study enrolled 122 human immunodeficiency virus (HIV) positive KS patients between 2017 and 2019 in Malawi. Participants were treated with bleomycin, vincristine and combination antiretroviral therapy, the local standard of care. One-year overall survival was 61%, and progression-free survival was 58%. The 48-week complete response rate was 35%. RNAseq (n = 78) differentiated two types of KS lesions, those with marked endothelial characteristics and those enriched in inflammatory transcripts. This suggests that different KS lesions are in different disease states consistent with the known heterogeneous clinical response to treatment. In contrast to earlier cohorts, the plasma HIV viral load of KS patients in our study was highly variable. A total of 25% of participants had no detectable HIV; all had detectable KSHV viral load. Our study affirms that many KS cases today develop in PLWH with well-controlled HIV infection and that different KS lesions have differing molecular compositions. Further studies are needed to develop predictive biomarkers for this disease.


Assuntos
Infecções por HIV , Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Sarcoma de Kaposi/complicações , Sarcoma de Kaposi/tratamento farmacológico , Sarcoma de Kaposi/epidemiologia , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Infecções por HIV/epidemiologia , HIV , Estudos Prospectivos , Herpesvirus Humano 8/fisiologia
11.
PLoS Pathog ; 17(1): e1009033, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411764

RESUMO

The p53 transcription factor plays a key role both in cancer and in the cell-intrinsic response to infections. The ORFEOME project hypothesized that novel p53-virus interactions reside in hitherto uncharacterized, unknown, or hypothetical open reading frames (orfs) of human viruses. Hence, 172 orfs of unknown function from the emerging viruses SARS-Coronavirus, MERS-Coronavirus, influenza, Ebola, Zika (ZIKV), Chikungunya and Kaposi Sarcoma-associated herpesvirus (KSHV) were de novo synthesized, validated and tested in a functional screen of p53 signaling. This screen revealed novel mechanisms of p53 virus interactions and two viral proteins KSHV orf10 and ZIKV NS2A binding to p53. Originally identified as the target of small DNA tumor viruses, these experiments reinforce the notion that all viruses, including RNA viruses, interfere with p53 functions. These results validate this resource for analogous systems biology approaches to identify functional properties of uncharacterized viral proteins, long non-coding RNAs and micro RNAs.


Assuntos
Doenças Transmissíveis Emergentes/virologia , Vírus de RNA/metabolismo , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/metabolismo , Vírus Chikungunya/genética , Vírus Chikungunya/metabolismo , Coronavirus/genética , Coronavirus/metabolismo , Ebolavirus/genética , Ebolavirus/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Fases de Leitura Aberta , Vírus de RNA/genética , Proteína Supressora de Tumor p53/genética , Proteínas não Estruturais Virais/metabolismo , Zika virus/genética , Zika virus/metabolismo
12.
J Med Virol ; 95(5): e28773, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37212317

RESUMO

This review will provide an overview of the notion that Kaposi sarcoma (KS) is a disease that manifests under diverse and divergent circumstances. We begin with a historical introduction of KS and KS-associated herpesvirus (KSHV), highlight the diversity of clinical presentations of KS, summarize what we know about the cell of origin for this tumor, explore KSHV viral load as a potential biomarker for acute KSHV infections and KS-associated complications, and discuss immune modulators that impact KSHV infection, KSHV persistence, and KS disease.


Assuntos
Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Carga Viral
13.
Immunity ; 40(3): 329-41, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24560620

RESUMO

Stimulator of interferon genes (STING, also named MITA, MYPS, or ERIS) is an intracellular DNA sensor that induces type I interferon through its interaction with TANK-binding kinase 1 (TBK1). Here we found that the nucleotide-binding, leucine-rich-repeat-containing protein, NLRC3, reduced STING-dependent innate immune activation in response to cytosolic DNA, cyclic di-GMP (c-di-GMP), and DNA viruses. NLRC3 associated with both STING and TBK1 and impeded STING-TBK1 interaction and downstream type I interferon production. By using purified recombinant proteins, we found NLRC3 to interact directly with STING. Furthermore, NLRC3 prevented proper trafficking of STING to perinuclear and punctated region, known to be important for its activation. In animals, herpes simplex virus 1 (HSV-1)-infected Nlrc3(-/-) mice exhibited enhanced innate immunity and reduced morbidity and viral load. This demonstrates the intersection of two key pathways of innate immune regulation, NLR and STING, to fine tune host response to intracellular DNA, DNA virus, and c-di-GMP.


Assuntos
DNA/imunologia , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Citocinas/biossíntese , Herpes Simples/imunologia , Herpes Simples/metabolismo , Herpesvirus Humano 1/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Interferon Tipo I/biossíntese , Camundongos , Camundongos Knockout , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico
14.
Proc Natl Acad Sci U S A ; 116(33): 16541-16550, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31346082

RESUMO

Non-Hodgkin lymphomas (NHLs) make up the majority of lymphoma diagnoses and represent a very diverse set of malignancies. We sought to identify kinases uniquely up-regulated in different NHL subtypes. Using multiplexed inhibitor bead-mass spectrometry (MIB/MS), we found Tyro3 was uniquely up-regulated and important for cell survival in primary effusion lymphoma (PEL), which is a viral lymphoma infected with Kaposi's sarcoma-associated herpesvirus (KSHV). Tyro3 was also highly expressed in PEL cell lines as well as in primary PEL exudates. Based on this discovery, we developed an inhibitor against Tyro3 named UNC3810A, which hindered cell growth in PEL, but not in other NHL subtypes where Tyro3 was not highly expressed. UNC3810A also significantly inhibited tumor progression in a PEL xenograft mouse model that was not seen in a non-PEL NHL model. Taken together, our data suggest Tyro3 is a therapeutic target for PEL.


Assuntos
Linfoma não Hodgkin/enzimologia , Linfoma de Efusão Primária/enzimologia , Terapia de Alvo Molecular , Proteoma/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/virologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
15.
J Virol ; 94(5)2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31801855

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of two B-cell lymphoproliferative diseases and Kaposi's sarcoma, an endothelial-cell-driven cancer. KSHV viral interleukin-6 (vIL-6) is a viral homolog of human IL-6 (hIL-6) that is expressed in KSHV-associated malignancies. Previous studies have shown that the expression of the integrin ß3 (ITGB3) subunit is induced upon KSHV infection. Here we report that KSHV vIL-6 is able to induce the expression of ITGB3 and increase surface expression of the αVß3 integrin heterodimer. We demonstrated using small interfering RNA (siRNA) depletion and inhibitor studies that KSHV vIL-6 can increase ITGB3 by inducing STAT3 signaling. Furthermore, we found that secreted vIL-6 is capable of inducing ITGB3 in endothelial cells in a paracrine manner. Importantly, the ability to induce ITGB3 in endothelial cells seems to be specific to vIL-6, as overexpression of hIL-6 alone did not affect levels of this integrin. Our lab and others have previously shown that vIL-6 can induce angiogenesis, and we investigated whether ITGB3 was involved in this process. We found that siRNA depletion of ITGB3 in vIL-6-expressing endothelial cells resulted in a decrease in adhesion to extracellular matrix proteins. Moreover, depletion of ITGB3 hindered the ability of vIL-6 to promote angiogenesis. In conclusion, we found that vIL-6 can singularly induce ITGB3 and that this induction is dependent on vIL-6 activation of the STAT3 signaling pathway.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of three human malignancies: multicentric Castleman's disease, primary effusion lymphoma, and Kaposi's sarcoma. Kaposi's sarcoma is a highly angiogenic tumor that arises from endothelial cells. It has been previously reported that KSHV infection of endothelial cells leads to an increase of integrin αVß3, a molecule observed to be involved in the angiogenic process of several malignancies. Our data demonstrate that the KSHV protein viral interleukin-6 (vIL-6) can induce integrin ß3 in an intracellular and paracrine manner. Furthermore, we showed that this induction is necessary for vIL-6-mediated cell adhesion and angiogenesis, suggesting a potential role of integrin ß3 in KSHV pathogenesis and development of Kaposi's sarcoma.


Assuntos
Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiologia , Integrina beta3/metabolismo , Interleucina-6/metabolismo , Fator de Transcrição STAT3/metabolismo , Sarcoma de Kaposi/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo , Hiperplasia do Linfonodo Gigante/virologia , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Humanos , Integrina beta3/genética , Linfoma de Efusão Primária/virologia , Sarcoma de Kaposi/virologia , Regulação para Cima
16.
PLoS Pathog ; 15(2): e1007536, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30716130

RESUMO

Extracellular signaling is a mechanism that higher eukaryotes have evolved to facilitate organismal homeostasis. Recent years have seen an emerging interest in the role of secreted microvesicles, termed extracellular vesicles (EV) or exosomes in this signaling network. EV contents can be modified by the cell in response to stimuli, allowing them to relay information to neighboring cells, influencing their physiology. Here we show that the tumor virus Kaposi's Sarcoma-associated herpesvirus (KSHV) hijacks this signaling pathway to induce cell proliferation, migration, and transcriptome reprogramming in cells not infected with the virus. KSHV-EV activates the canonical MEK/ERK pathway, while not alerting innate immune regulators, allowing the virus to exert these changes without cellular pathogen recognition. Collectively, we propose that KSHV establishes a niche favorable for viral spread and cell transformation through cell-derived vesicles, all while avoiding detection.


Assuntos
Reprogramação Celular/fisiologia , Vesículas Extracelulares/fisiologia , Herpesvirus Humano 8/metabolismo , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Reprogramação Celular/genética , Células Endoteliais/fisiologia , Herpesvirus Humano 8/genética , Interações Hospedeiro-Patógeno , Células Endoteliais da Veia Umbilical Humana , Humanos , Linfoma/genética , Linfoma/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Transdução de Sinais , Transcriptoma/genética , Proteínas Virais , Latência Viral
17.
Immunity ; 36(6): 933-46, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22749352

RESUMO

The mitochondrial protein MAVS (also known as IPS-1, VISA, and CARDIF) interacts with RIG-I-like receptors (RLRs) to induce type I interferon (IFN-I). NLRX1 is a mitochondrial nucleotide-binding, leucine-rich repeats (NLR)-containing protein that attenuates MAVS-RLR signaling. Using Nlrx1(-/-) cells, we confirmed that NLRX1 attenuated IFN-I production, but additionally promoted autophagy during viral infection. This dual function of NLRX1 paralleled the previously described functions of the autophagy-related proteins Atg5-Atg12, but NLRX1 did not associate with Atg5-Atg12. High-throughput quantitative mass spectrometry and endogenous protein-protein interaction revealed an NLRX1-interacting partner, mitochondrial Tu translation elongation factor (TUFM). TUFM interacted with Atg5-Atg12 and Atg16L1 and has similar functions as NLRX1 by inhibiting RLR-induced IFN-I but promoting autophagy. In the absence of NLRX1, increased IFN-I and decreased autophagy provide an advantage for host defense against vesicular stomatitis virus. This study establishes a link between an NLR protein and the viral-induced autophagic machinery via an intermediary partner, TUFM.


Assuntos
Autofagia/fisiologia , Interferon Tipo I/biossíntese , Proteínas Mitocondriais/fisiologia , Fator Tu de Elongação de Peptídeos/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Sequência de Aminoácidos , Animais , Proteína 12 Relacionada à Autofagia , Proteína 5 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/fisiologia , Citocinas/biossíntese , Citocinas/genética , Proteína DEAD-box 58 , RNA Helicases DEAD-box/fisiologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica/imunologia , Células HEK293 , Humanos , Interferon Tipo I/genética , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/imunologia , Camundongos , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Mitocondriais/química , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Dados de Sequência Molecular , Complexos Multiproteicos/fisiologia , Fator Tu de Elongação de Peptídeos/química , Mapeamento de Interação de Proteínas , Proteínas/fisiologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/fisiologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Organismos Livres de Patógenos Específicos , Vesiculovirus/fisiologia
18.
J Virol ; 93(11)2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30867309

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human oncogenic virus. KSHV utilizes its proteins to modify the cellular environment to promote viral replication and persistence. Some of these proteins are oncogenic, modulating cell proliferation, apoptosis, angiogenesis, genome stability, and immune responses, among other cancer hallmarks. These changes can lead to the development of KSHV-associated malignancies. In this Gem, we focus on animal models of oncogenic KSHV proteins that were developed to enable better understanding of KSHV tumorigenesis.


Assuntos
Herpesvirus Humano 8/metabolismo , Proteínas Oncogênicas/metabolismo , Animais , Carcinogênese , Modelos Animais de Doenças , Herpesvirus Humano 8/genética , Humanos , Camundongos , Proteínas Oncogênicas/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/metabolismo , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa