Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Med Virol ; 95(8): e29009, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37563850

RESUMO

Despite intensive studies during the last 3 years, the pathology and underlying molecular mechanism of coronavirus disease 2019 (COVID-19) remain poorly defined. In this study, we investigated the spatial single-cell molecular and cellular features of postmortem COVID-19 lung tissues using in situ sequencing (ISS). We detected 10 414 863 transcripts of 221 genes in whole-slide tissues and segmented them into 1 719 459 cells that were mapped to 18 major parenchymal and immune cell types, all of which were infected by SARS-CoV-2. Compared with the non-COVID-19 control, COVID-19 lungs exhibited reduced alveolar cells (ACs) and increased innate and adaptive immune cells. We also identified 19 differentially expressed genes in both infected and uninfected cells across the tissues, which reflected the altered cellular compositions. Spatial analysis of local infection rates revealed regions with high infection rates that were correlated with high cell densities (HIHD). The HIHD regions expressed high levels of SARS-CoV-2 entry-related factors including ACE2, FURIN, TMPRSS2 and NRP1, and co-localized with organizing pneumonia (OP) and lymphocytic and immune infiltration, which exhibited increased ACs and fibroblasts but decreased vascular endothelial cells and epithelial cells, mirroring the tissue damage and wound healing processes. Sparse nonnegative matrix factorization (SNMF) analysis of niche features identified seven signatures that captured structure and immune niches in COVID-19 tissues. Trajectory inference based on immune niche signatures defined two pathological routes. Trajectory A primarily progressed with increased NK cells and granulocytes, likely reflecting the complication of microbial infections. Trajectory B was marked by increased HIHD and OP, possibly accounting for the increased immune infiltration. The OP regions were marked by high numbers of fibroblasts expressing extremely high levels of COL1A1 and COL1A2. Examination of single-cell RNA-seq data (scRNA-seq) from COVID-19 lung tissues and idiopathic pulmonary fibrosis (IPF) identified similar cell populations consisting mainly of myofibroblasts. Immunofluorescence staining revealed the activation of IL6-STAT3 and TGF-ß-SMAD2/3 pathways in these cells, likely mediating the upregulation of COL1A1 and COL1A2 and excessive fibrosis in the lung tissues. Together, this study provides a spatial single-cell atlas of cellular and molecular signatures of fatal COVID-19 lungs, which reveals the complex spatial cellular heterogeneity, organization, and interactions that characterized the COVID-19 lung pathology.


Assuntos
COVID-19 , Humanos , COVID-19/patologia , SARS-CoV-2/genética , Células Endoteliais , Análise da Expressão Gênica de Célula Única , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Pulmão/patologia
2.
J Infect Dis ; 223(11): 1842-1854, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33837392

RESUMO

BACKGROUND: Coronavirus disease 2019 (COVID-19) patients manifest with pulmonary symptoms reflected by diffuse alveolar damage (DAD), excessive inflammation, and thromboembolism. The mechanisms mediating these processes remain unclear. METHODS: We performed multicolor staining for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) proteins and lineage markers to define viral tropism and lung pathobiology in 5 autopsy cases. RESULTS: Lung parenchyma showed severe DAD with thromboemboli. Viral infection was found in an extensive range of cells including pneumocyte type II, ciliated, goblet, club-like, and endothelial cells. More than 90% of infiltrating immune cells were positive for viral proteins including macrophages, monocytes, neutrophils, natural killer (NK) cells, B cells, and T cells. Most but not all infected cells were angiotensin-converting enzyme 2 (ACE2) positive. The numbers of infected and ACE2-positive cells are associated with extensive tissue damage. Infected tissues exhibited high levels of inflammatory cells including macrophages, monocytes, neutrophils, and NK cells, and low levels of B cells but abundant T cells consisting of mainly T helper cells, few cytotoxic T cells, and no regulatory T cells. Robust interleukin-6 expression was present in most cells, with or without infection. CONCLUSIONS: In fatal COVID-19 lungs, there are broad SARS-CoV-2 cell tropisms, extensive infiltrated innate immune cells, and activation and depletion of adaptive immune cells, contributing to severe tissue damage, thromboemboli, excess inflammation, and compromised immune responses.


Assuntos
COVID-19/patologia , Pulmão/patologia , SARS-CoV-2/fisiologia , Tropismo Viral , Adulto , Idoso , COVID-19/imunologia , COVID-19/virologia , Feminino , Humanos , Imunidade Inata , Pulmão/citologia , Pulmão/imunologia , Pulmão/virologia , Masculino , Pessoa de Meia-Idade , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/patologia , Alvéolos Pulmonares/virologia , Tropismo Viral/imunologia
3.
J Med Virol ; 93(8): 5065-5075, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33942339

RESUMO

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


Assuntos
Antígenos Virais/genética , Sistemas CRISPR-Cas , Transformação Celular Neoplásica/genética , Herpesvirus Humano 8/genética , Proteínas Nucleares/genética , Vírus Oncogênicos/genética , Animais , Antígenos Virais/metabolismo , Apoptose , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Ciclo Celular , Proliferação de Células , Técnicas de Inativação de Genes , Genoma Viral/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Linfoma de Efusão Primária/patologia , Células-Tronco Mesenquimais , Proteínas Nucleares/metabolismo , Vírus Oncogênicos/patogenicidade , RNA Guia de Cinetoplastídeos/genética , Ratos , Latência Viral/genética
4.
J Med Virol ; 93(12): 6671-6685, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34324210

RESUMO

Infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a wide spectrum of syndromes involving multiple organ systems and is primarily mediated by viral spike (S) glycoprotein through the receptor-binding domain (RBD) and numerous cellular proteins including ACE2, transmembrane serine protease 2 (TMPRSS2), kidney injury molecule-1 (Kim-1), and neuropilin-1 (NRP-1). In this study, we examined the entry tropism of SARS-CoV-2 and SARS-CoV using S protein-based pseudoviruses to infect 22 cell lines and 3 types of primary cells isolated from respiratory, urinary, digestive, reproductive, and immune systems. At least one cell line or type of primary cell from each organ system was infected by both pseudoviruses. Infection by pseudoviruses is effectively blocked by S1, RBD, and ACE2 recombinant proteins, and more weakly by Kim-1 and NRP-1 recombinant proteins. Furthermore, cells with robust SARS-CoV-2 pseudovirus infection had strong expression of either ACE2 or Kim-1 and NRP-1 proteins. ACE2 glycosylation appeared to be critical for the infections of both viruses as there was a positive correlation between infectivity of either SARS-CoV-2 or SARS-CoV pseudovirus with the level of glycosylated ACE2 (gly-ACE2). These results reveal that SARS-CoV-2 cell entry could be mediated by either an ACE2-dependent or -independent mechanism, thus providing a likely molecular basis for its broad tropism for a wide variety of cell types.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Trato Gastrointestinal/virologia , Genitália/virologia , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Sistema Imunitário/virologia , Neuropilina-1/metabolismo , Sistema Respiratório/virologia , SARS-CoV-2/fisiologia , Serina Endopeptidases/metabolismo , Internalização do Vírus , Western Blotting , COVID-19/metabolismo , COVID-19/virologia , Linhagem Celular , Células Cultivadas , Imunofluorescência , Trato Gastrointestinal/citologia , Genitália/citologia , Humanos , Sistema Imunitário/citologia , Sistema Respiratório/citologia
5.
J Virol ; 93(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30404794

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) has latent and lytic replication phases, both of which contribute to the development of KSHV-induced malignancies. Among the numerous factors identified to regulate the KSHV life cycle, oxidative stress, caused by imbalanced clearing and production of reactive oxygen species (ROS), has been shown to robustly disrupt KSHV latency and induce viral lytic replication. In this study, we identified an important role of the antioxidant defense factor forkhead box protein O1 (FoxO1) in the KSHV life cycle. Either chemical inhibition of the FoxO1 function or knockdown of FoxO1 expression led to an increase in the intracellular ROS level that was subsequently sufficient to disrupt KSHV latency and induce viral lytic reactivation. On the other hand, treatment with N-acetyl-l-cysteine (NAC), an oxygen free radical scavenger, led to a reduction in the FoxO1 inhibition-induced ROS level and, ultimately, the attenuation of KSHV lytic reactivation. These findings reveal that FoxO1 plays a critical role in keeping KSHV latency in check by maintaining the intracellular redox balance.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with several cancers, including Kaposi's sarcoma (KS). Both the KSHV latent and lytic replication phases are important for the development of KS. Identification of factors regulating the KSHV latent phase-to-lytic phase switch can provide insights into the pathogenesis of KSHV-induced malignancies. In this study, we show that the antioxidant defense factor forkhead box protein O1 (FoxO1) maintains KSHV latency by suppressing viral lytic replication. Inhibition of FoxO1 disrupts KSHV latency and induces viral lytic replication by increasing the intracellular ROS level. Significantly, treatment with an oxygen free radical scavenger, N-acetyl-l-cysteine (NAC), attenuated the FoxO1 inhibition-induced intracellular ROS level and KSHV lytic replication. Our works reveal a critical role of FoxO1 in suppressing KSHV lytic replication, which could be targeted for antiviral therapy.


Assuntos
Proteína Forkhead Box O1/metabolismo , Herpesvirus Humano 8/fisiologia , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Ativação Viral , Latência Viral , Replicação Viral , Células Cultivadas , Proteína Forkhead Box O1/antagonistas & inibidores , Proteína Forkhead Box O1/genética , Humanos , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Sarcoma de Kaposi/genética
6.
J Virol ; 92(4)2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29212931

RESUMO

The recent outbreak of Zika virus (ZIKV), a reemerging flavivirus, and its associated neurological disorders, such as Guillain-Barré (GB) syndrome and microcephaly, have generated an urgent need to develop effective ZIKV vaccines and therapeutic agents. Here, we used human endothelial cells and astrocytes, both of which represent key cell types for ZIKV infection, to identify potential inhibitors of ZIKV replication. Because several pathways, including the AMP-activated protein kinase (AMPK), protein kinase A (PKA), and mitogen-activated protein kinase (MAPK) signaling pathways, have been reported to play important roles in flavivirus replication, we tested inhibitors and agonists of these pathways for their effects on ZIKV replication. We identified the PKA inhibitor PKI 14-22 (PKI) to be a potent inhibitor of ZIKV replication. PKI effectively suppressed the replication of ZIKV from both the African and Asian/American lineages with a high efficiency and minimal cytotoxicity. While ZIKV infection does not induce PKA activation, endogenous PKA activity is essential for supporting ZIKV replication. Interestingly, in addition to PKA, PKI also inhibited another unknown target(s) to block ZIKV replication. PKI inhibited ZIKV replication at the postentry stage by preferentially affecting negative-sense RNA synthesis as well as viral protein translation. Together, these results have identified a potential inhibitor of ZIKV replication which could be further explored for future therapeutic application.IMPORTANCE There is an urgent need to develop effective vaccines and therapeutic agents against Zika virus (ZIKV) infection, a reemerging flavivirus associated with neurological disorders, including Guillain-Barré (GB) syndrome and microcephaly. By screening for inhibitors of several cellular pathways, we have identified the PKA inhibitor PKI 14-22 (PKI) to be a potent inhibitor of ZIKV replication. We show that PKI effectively suppresses the replication of all ZIKV strains tested with minimal cytotoxicity to human endothelial cells and astrocytes, two key cell types for ZIKV infection. Furthermore, we show that PKI inhibits ZIKV negative-sense RNA synthesis and viral protein translation. This study has identified a potent inhibitor of ZIKV infection which could be further explored for future therapeutic application.


Assuntos
Astrócitos/efeitos dos fármacos , Proteínas de Transporte/farmacologia , Células Endoteliais/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Fragmentos de Peptídeos/farmacologia , Zika virus/efeitos dos fármacos , Animais , Astrócitos/virologia , Chlorocebus aethiops , Células Endoteliais/virologia , Humanos , Sistema de Sinalização das MAP Quinases , Células Vero , Replicação Viral/efeitos dos fármacos , Infecção por Zika virus/tratamento farmacológico
7.
J Med Virol ; 91(2): 179-189, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30192399

RESUMO

After recent outbreaks, Zika virus (ZIKV) was linked to severe neurological diseases including Guillain-Barré syndrome in adults and microcephaly in newborns. The severities of pathological manifestations have been associated with different ZIKV strains. To better understand the tropism of ZIKV, we infected 10 human and four nonhuman cell lines (types) with two African (IbH30656 and MR766) and two Asian (PRVABC59 and H/FP/2013) ZIKV strains. Cell susceptibility to ZIKV infection was determined by examining viral titers, synthesis of viral proteins, and replication of positive and negative strands of viral genome. Among nonhuman cell lines, only Vero cells were efficiently infected by ZIKV. Among human cell lines, all were permissive to ZIKV infection. However, 293T and HeLa cells showed differential susceptibility towards African strains. In 293T cells, the NS1 protein was expressed at the high level by African strains but was almost not expressed by Asian strains though there was no obvious difference in viral genome replication, suggesting that the differential susceptibility might be controlled at the stage of viral protein translation. This study provides comprehensive results of the permissiveness of different cell types to both African and Asian ZIKV strains, which might help clarify their different pathogenesis.


Assuntos
Tropismo Viral , Replicação Viral , Zika virus/crescimento & desenvolvimento , Animais , Linhagem Celular , Humanos , Zika virus/isolamento & purificação , Infecção por Zika virus/virologia
8.
PLoS Pathog ; 12(5): e1005648, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27187079

RESUMO

Aerobic glycolysis is essential for supporting the fast growth of a variety of cancers. However, its role in the survival of cancer cells under stress conditions is unclear. We have previously reported an efficient model of gammaherpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV)-induced cellular transformation of rat primary mesenchymal stem cells. KSHV-transformed cells efficiently induce tumors in nude mice with pathological features reminiscent of Kaposi's sarcoma tumors. Here, we report that KSHV promotes cell survival and cellular transformation by suppressing aerobic glycolysis and oxidative phosphorylation under nutrient stress. Specifically, KSHV microRNAs and vFLIP suppress glycolysis by activating the NF-κB pathway to downregulate glucose transporters GLUT1 and GLUT3. While overexpression of the transporters rescues the glycolytic activity, it induces apoptosis and reduces colony formation efficiency in softagar under glucose deprivation. Mechanistically, GLUT1 and GLUT3 inhibit constitutive activation of the AKT and NF-κB pro-survival pathways. Strikingly, GLUT1 and GLUT3 are significantly downregulated in KSHV-infected cells in human KS tumors. Furthermore, we have detected reduced levels of aerobic glycolysis in several KSHV-infected primary effusion lymphoma cell lines compared to a Burkitt's lymphoma cell line BJAB, and KSHV infection of BJAB cells reduced aerobic glycolysis. These results reveal a novel mechanism by which an oncogenic virus regulates a key metabolic pathway to adapt to stress in tumor microenvironment, and illustrate the importance of fine-tuning the metabolic pathways for sustaining the proliferation and survival of cancer cells, particularly under stress conditions.


Assuntos
Adaptação Fisiológica/fisiologia , Transformação Celular Viral/fisiologia , Infecções por Herpesviridae/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular , Modelos Animais de Doenças , Citometria de Fluxo , Imunofluorescência , Técnicas de Silenciamento de Genes , Glicólise/fisiologia , Herpesvirus Humano 8/metabolismo , Humanos , Linfoma de Efusão Primária/metabolismo , Linfoma de Efusão Primária/virologia , Microscopia Confocal , Reação em Cadeia da Polimerase , Ratos
9.
J Pathol ; 242(3): 309-321, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28393364

RESUMO

Primary effusion lymphoma (PEL) is a rare and aggressive B-cell lymphoma with a dismal prognosis caused by infection of Kaposi's sarcoma-associated herpesvirus. Despite the findings that numerous viral genes and cellular pathways are essential for the proliferation and survival of PEL cells, there is currently no effective therapeutic treatment for PEL. Here, we report that the metabolic sensor SIRT1 is functionally required for sustaining the proliferation and survival of PEL cells. Knockdown of SIRT1 with specific shRNAs or inhibition of SIRT1 with an inhibitor (tenovin-6) induced cell cycle arrest and apoptosis in PEL cells. We detected high levels of AMPK activation in PEL cells, reflected in AMPKα1 phosphorylation at T174. Knockdown or inhibition of SIRT1 reduced AMPK activation, indicating that SIRT1 was required for AMPK activation. Interestingly, knockdown of AMPK with specific shRNAs or inhibition of AMPK with the inhibitor compound C recapitulated the phenotype of SIRT1, and induced cell cycle arrest and apoptosis, whereas overexpression of a constitutively active AMPK construct rescued the cytotoxic effect of SIRT1 knockdown. Remarkably, treatment with tenovin-6 effectively inhibited the initiation and progression of PEL, and significantly extended the survival of mice in a murine PEL model. Taken together, these results illustrate that the SIRT1-AMPK axis is essential for maintaining the proliferation and survival of PEL and identify SIRT1 and AMPK as potential therapeutic targets, and tenovin-6 as a candidate therapeutic agent for PEL patients. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Linfoma de Efusão Primária/fisiopatologia , Sirtuína 1/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/fisiologia , Benzamidas/farmacologia , Pontos de Checagem do Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Técnicas de Silenciamento de Genes , Linfoma de Efusão Primária/enzimologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos Endogâmicos NOD , Camundongos SCID , Fosforilação/fisiologia , Sirtuína 1/antagonistas & inibidores
10.
Proc Natl Acad Sci U S A ; 111(41): E4342-9, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25271323

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr virus (EBV) are human DNA tumor viruses that express nuclear antigens [latency-associated nuclear antigen 1 (LANA1) and Epstein-Barr nuclear antigen 1 (EBNA1)] necessary to maintain and replicate the viral genome. We report here that both LANA1 and EBNA1 undergo highly efficient +1/-2 programmed ribosomal frameshifting to generate previously undescribed alternative reading frame (ARF) proteins in their repeat regions. EBNA1(ARF) encodes a KSHV LANA-like glutamine- and glutamic acid-rich protein, whereas KSHV LANA1(ARF) encodes a serine/arginine-like protein. Repeat sequence recoding has not been described previously for human DNA viruses. Programmed frameshifting (recoding) to generate multiple proteins from one RNA sequence can increase the coding capacity of a virus, without incurring a selective penalty against increased capsid size. The presence of similar repeat sequences in cellular genes, such as huntingtin, suggests that a comparison of repeat recoding in virus and human systems may provide functional and mechanistic insights for both systems.


Assuntos
Antígenos Virais/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/metabolismo , Fases de Leitura/genética , Sequências Repetitivas de Aminoácidos , Sequência de Aminoácidos , Antígenos Virais/química , Linhagem Celular Tumoral , Citoplasma/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/química , Mudança da Fase de Leitura do Gene Ribossômico , Células HEK293 , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/química , Iniciação Traducional da Cadeia Peptídica , Estrutura Terciária de Proteína
11.
J Med Virol ; 88(8): 1291-6, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27124623

RESUMO

Zika virus (ZIKV) was first described in 1947, and became a health emergency problem in 2016 when its association with fetal microcephaly cases was confirmed by Centers for Disease Control and Prevention (CDC) in the United States. To date, ZIKV infection has been documented in 66 countries. ZIKV is recognized as a neurotropic virus and numerous diseases manifested in multiple neurological disorders have been described, mainly in countries that have been exposed to ZIKV after the 2007 outbreak in the Federated States of Micronesia. The most dramatic consequence of ZIKV infection documented is the abrupt increase in fetal microcephaly cases in Brazil. Here, we present an update of the published research progress in the past few months. J. Med. Virol. 88:1291-1296, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Modelos Animais de Doenças , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/fisiopatologia , Zika virus/genética , Aedes/virologia , Animais , Brasil/epidemiologia , Surtos de Doenças , Humanos , Insetos Vetores/virologia , Camundongos , Microcefalia/virologia , Micronésia/epidemiologia , Doenças do Sistema Nervoso/virologia , Primatas , Infecção por Zika virus/virologia
12.
J Med Virol ; 88(10): 1657-8, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27208545

RESUMO

Three recent studies in pregnant mice and one ongoing study in rhesus macaques evaluating the effect of ZIKV infection have provided important information about maternal-fetus transmission and ZIKV-related pathogenesis, confirming a causal role of ZIKV in neurological problems observed in humans. Here, we present an update of these works published in the past few weeks. J. Med. Virol. 88:1657-1658, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Modelos Animais de Doenças , Transmissão Vertical de Doenças Infecciosas , Complicações Infecciosas na Gravidez/virologia , Infecção por Zika virus/transmissão , Infecção por Zika virus/virologia , Zika virus/patogenicidade , Animais , Feminino , Humanos , Macaca mulatta , Camundongos , Gravidez , Infecção por Zika virus/complicações
13.
J Med Virol ; 85(12): 2110-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24014234

RESUMO

Patients infected with the human immunodeficiency virus (HIV) are at higher risk of developing Epstein-Barr Virus (EBV)-associated lymphomas. The usefulness of monitoring EBV in peripheral blood mononuclear cells (PBMCs) of patients infected with HIV has not been established. The aim of this study was to evaluate the EBV viral load in PBMCs, the frequency of viral genotypes, and the presence of the 30-bp deletion in the BNLF-1 gene. DNA samples from 156 patients attending the HIV/AIDS Day Clinic at Botucatu School of Medicine, Sao Paulo State University were evaluated. The EBV viral load was detectable by real time PCR in 123/156 (78.8%) cases and was higher in patients not receiving antiretroviral treatment or under therapeutic failure than in patients under successful highly active antiretroviral therapy (HAART) (P = 0.0076). Overall, the profile of patients with high EBV viral load included elevated HIV viremia (P = 0.0005), longer time of HIV diagnosis (P = 0.0026), and increased levels of T CD8 (+) lymphocytes (P = 0.0159). The successful amplification of the EBNA-2 gene by nested-PCR was achieved in 95 of 123 (77.2%) cases, of which 75.8% were EBV-1, 9.5% EBV-2, and 14.7% were co-infected with both EBV-1 and -2. The analysis of the BNLF-1 gene was possible in 99 of 123 (80.5%) cases, of which 50.5% had the 30-bp deletion. EBV-1 was more common than EBV-2, which may reflect the fact that the cohort was predominantly Caucasian and heterosexual.


Assuntos
Coinfecção , Infecções por Vírus Epstein-Barr/virologia , Genótipo , Infecções por HIV , Herpesvirus Humano 4/genética , Deleção de Sequência , Proteínas da Matriz Viral/genética , Terapia Antirretroviral de Alta Atividade , Contagem de Linfócito CD4 , Feminino , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Leucócitos Mononucleares/virologia , Masculino , Carga Viral
14.
ACS Appl Mater Interfaces ; 11(38): 34717-34724, 2019 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-31469541

RESUMO

The CRISPR/Cas gene editing system has been successfully applied to combating bacteria, cancer, virus, and genetic disorders. While viral vectors have been used for the delivery of the CRISPR/Cas9 system, the time required for insert cloning, and virus packaging and standardization, hinders its efficient use. Additionally, the high molecular weight of the Cas9 endonuclease makes it not easy for packing into the vehicles. Herein we report the self-assembly of gold nanoclusters (AuNCs) with SpCas9 protein (SpCas9-AuNCs) under physiological conditions and the efficient delivery of SpCas9 into the cell nucleus. This assembly process is highly dependent on pH. SpCas9-AuNCs are stable at a higher pH but are disassembled at a lower pH. Significantly, this assembly-disassembly process facilitates the delivery of SpCas9 into cells and the cell nucleus, where the SpCas9 exerts its cleavage function. As a proof-of-concept, the assembled SpCas9-AuNCs nanoparticles are successfully used for efficient knockout of the E6 oncogene, restoring the function of tumor-suppressive protein p53 and inducing apoptosis in cervical cancer cells with little effect on normal human cells. The SpCas9-AuNCs are useful for sgRNA functional validation, sgRNA library screening, and genomic manipulation.


Assuntos
Proteína 9 Associada à CRISPR , Genes Virais , Ouro , Nanopartículas Metálicas , Oncogenes , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero , Proteína 9 Associada à CRISPR/química , Proteína 9 Associada à CRISPR/farmacologia , Feminino , Ouro/química , Ouro/farmacologia , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/virologia
15.
Am J Clin Pathol ; 128(5): 794-802, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17951202

RESUMO

We aimed to evaluate the frequency of Kaposi sarcoma (KS)-associated herpesvirus (KSHV) infection in KS lesions in patients from Brazil. In addition, expression of human bcl-2, cleaved caspase-3, and KSHV latency-associated nuclear antigen (LANA)-1 in tumors was evaluated using immunohistochemical analysis. We studied 64 KS cases, classified as follows: classical, 20 (31%); iatrogenic, 2 (3%); AIDS-associated, 25 (39%); and not otherwise specified (lack of information about HIV status), 17 (27%). KSHV was detected by polymerase chain reaction (PCR) in 61 cases (95%); 40 cases (63%) were KSHV+ by PCR and immunohistochemical analysis for LANA-1. Immunoexpression of bcl-2 was detected in 47 cases (73%). Only a few cells in 15 cases (23%) of KS had demonstrable immunostaining for cleaved caspase-3. These results further support the association of KSHV with all KS forms. Cleaved caspase-3 in KS tumors was infrequent, which may reflect the inhibition of apoptosis owing to bcl-2 overexpression observed in the majority of KS tumors.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/metabolismo , Antígenos Virais/metabolismo , Caspase 3/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sarcoma de Kaposi/metabolismo , Neoplasias Cutâneas/metabolismo , Proteínas Virais/metabolismo , Infecções Oportunistas Relacionadas com a AIDS/patologia , Infecções Oportunistas Relacionadas com a AIDS/virologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Brasil , DNA Viral/análise , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/isolamento & purificação , Humanos , Masculino , Pessoa de Meia-Idade , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/virologia
16.
Cancer Res ; 77(24): 7094-7108, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29051178

RESUMO

Toll-like receptors (TLR) are conserved immune sensors mediating antimicrobial and antitumoral responses, but recent evidence implicates them in promoting carcinogenesis in certain cancers. Kaposi sarcoma is caused by infection of Kaposi sarcoma-associated herpesvirus (KSHV) and is characterized by uncontrolled neoangiogenesis and inflammation. Here, we show that TLR4 is upregulated in KSHV-infected spindle tumor cells in human Kaposi sarcoma lesions. In a model of KSHV-induced cellular transformation, KSHV upregulated expression of TLR4, its adaptor MyD88, and coreceptors CD14 and MD2. KSHV induction of TLR4 was mediated by multiple viral miRNAs. Importantly, the TLR4 pathway was activated constitutively in KSHV-transformed cells, resulting in chronic induction of IL6, IL1ß, and IL18. Accordingly, IL6 mediated constitutive activation of the STAT3 pathway, an essential event for uncontrolled cellular proliferation and transformation. TLR4 stimulation with lipopolysaccharides or live bacteria enhanced tumorigenesis while TLR4 antagonist CLI095 inhibited it. These results highlight an essential role of the TLR4 pathway and chronic inflammation in KSHV-induced tumorigenesis, which helps explain why HIV-infected patients, who frequently suffer from opportunistic bacterial infections and metabolic complications, frequently develop Kaposi sarcoma. Cancer Res; 77(24); 7094-108. ©2017 AACR.


Assuntos
Transformação Celular Viral/genética , Herpesvirus Humano 8/fisiologia , Inflamação/complicações , Sarcoma de Kaposi/genética , Receptor 4 Toll-Like/fisiologia , Animais , Carcinogênese/genética , Proliferação de Células/genética , Células Cultivadas , Feminino , Humanos , Inflamação/genética , Mediadores da Inflamação/fisiologia , Camundongos , Camundongos Nus , Ratos , Fator de Transcrição STAT3/metabolismo , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Transdução de Sinais/genética
17.
mBio ; 8(4)2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28811348

RESUMO

While glutamine is a nonessential amino acid that can be synthesized from glucose, some cancer cells primarily depend on glutamine for their growth, proliferation, and survival. Numerous types of cancer also depend on asparagine for cell proliferation. The underlying mechanisms of the glutamine and asparagine requirement in cancer cells in different contexts remain unclear. In this study, we show that the oncogenic virus Kaposi's sarcoma-associated herpesvirus (KSHV) accelerates the glutamine metabolism of glucose-independent proliferation of cancer cells by upregulating the expression of numerous critical enzymes, including glutaminase 2 (GLS2), glutamate dehydrogenase 1 (GLUD1), and glutamic-oxaloacetic transaminase 2 (GOT2), to support cell proliferation. Surprisingly, cell crisis is rescued only completely by supplementation with asparagine but minimally by supplementation with α-ketoglutarate, aspartate, or glutamate upon glutamine deprivation, implying an essential role of γ-nitrogen in glutamine and asparagine for cell proliferation. Specifically, glutamine and asparagine provide the critical γ-nitrogen for purine and pyrimidine biosynthesis, as knockdown of four rate-limiting enzymes in the pathways, including carbamoylphosphate synthetase 2 (CAD), phosphoribosyl pyrophosphate amidotransferase (PPAT), and phosphoribosyl pyrophosphate synthetases 1 and 2 (PRPS1 and PRPS2, respectively), suppresses cell proliferation. These findings indicate that glutamine and asparagine are shunted to the biosynthesis of nucleotides and nonessential amino acids from the tricarboxylic acid (TCA) cycle to support the anabolic proliferation of KSHV-transformed cells. Our results illustrate a novel mechanism by which an oncogenic virus hijacks a metabolic pathway for cell proliferation and imply potential therapeutic applications in specific types of cancer that depend on this pathway.IMPORTANCE We have previously found that Kaposi's sarcoma-associated herpesvirus (KSHV) can efficiently infect and transform primary mesenchymal stem cells; however, the metabolic pathways supporting the anabolic proliferation of KSHV-transformed cells remain unknown. Glutamine and asparagine are essential for supporting the growth, proliferation, and survival of some cancer cells. In this study, we have found that KSHV accelerates glutamine metabolism by upregulating numerous critical metabolic enzymes. Unlike most cancer cells that primarily utilize glutamine and asparagine to replenish the TCA cycle, KSHV-transformed cells depend on glutamine and asparagine for providing γ-nitrogen for purine and pyrimidine biosynthesis. We identified four rate-limiting enzymes in this pathway that are essential for the proliferation of KSHV-transformed cells. Our results demonstrate a novel mechanism by which an oncogenic virus hijacks a metabolic pathway for cell proliferation and imply potential therapeutic applications in specific types of cancer that depend on this pathway.


Assuntos
Asparagina/metabolismo , Proliferação de Células , Glutamina/metabolismo , Herpesvirus Humano 8/fisiologia , Neoplasias/patologia , Neoplasias/virologia , Nucleotídeos/biossíntese , Asparagina/farmacologia , Aspartato Aminotransferases/genética , Ácido Aspártico/farmacologia , Proliferação de Células/efeitos dos fármacos , Glutamato Desidrogenase/genética , Ácido Glutâmico/farmacologia , Glutaminase/genética , Glutamina/deficiência , Humanos , Redes e Vias Metabólicas , Neoplasias/fisiopatologia , Nitrogênio/metabolismo
18.
J Clin Pathol ; 68(9): 726-32, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25979989

RESUMO

BACKGROUND: The Epstein-Barr virus (EBV) and Kaposi's sarcoma associated herpesvirus (KSHV) are consistently associated with lymphoproliferative diseases and cancers in humans, notably in patients with HIV. AIMS: Our aim was to evaluate whether EBV and/or KSHV viral loads regularly assessed in peripheral blood mononuclear cells (PBMC) correlate with clinical or laboratorial parameters retrieved for patients living with HIV. METHODS: This was a longitudinal study with a cohort of 157 HIV positive patients attending an academic HIV outpatient clinic in São Paulo State, Brazil. For each patient, up to four blood samples were collected over a 1 year clinical follow-up: on enrolment into the study, and after 4, 8 and 12 months. Total DNA was extracted from PBMC, and EBV and KSHV viral loads were assessed by real time quantitative PCR. RESULTS: Higher viral loads for EBV were significantly associated with high HIV viraemia, a greater number of circulating T CD8+ cells and lack of virological response to the antiretroviral treatment. KSHV viral load was undetectable in virtually all samples. CONCLUSIONS: EBV viral load in PBMC correlated with the number of circulating T CD8+ lymphocytes and the response to the antiretroviral therapy in HIV infected patients. In contrast, KSHV was undetectable in PBMC, presumably an effect of the antiretroviral treatment. Therefore, either KSHV infection in the population studied was absent or viral load in PBMC was beyond the analytical limit of the assay.


Assuntos
Coinfecção , Infecções por HIV/complicações , Infecções por HIV/virologia , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/virologia , Viremia/complicações , Adolescente , Adulto , Idoso , Estudos de Coortes , Ensaio de Imunoadsorção Enzimática , Feminino , Herpesvirus Humano 4 , Herpesvirus Humano 8 , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Adulto Jovem
19.
Cell Cycle ; 13(5): 845-58, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24419204

RESUMO

Kaposi sarcoma-associated herpesvirus (KSHV) is a tumor virus encoding several proto-oncogenes. However, the roles of these viral genes in KSHV-induced tumorigenesis have not been defined. In this study, we used a recently developed model of KSHV-induced cellular transformation and tumorigenesis combining with a reverse genetic system to examine the role of a KSHV latent gene vCyclin (ORF72), a cellular Cyclin D2 homolog, in KSHV-induced oncogenesis. Deletion of vCyclin did not affect cell proliferation and cell cycle progression at a low-density condition, when cells were at an active proliferation state. However, vCyclin mutant cells were contact-inhibited and arrested at G 1 phase at a high-density condition. As a result, vCyclin mutant cells formed less and smaller colonies in soft agar assay. Nude mice inoculated with vCyclin mutant cells had reduced tumor incidence and extended tumor latency and survival compared with mice inoculated with wild-type (WT) virus-infected cells. WT but not mutant virus effectively induced Cyclin-dependent kinase inhibitor p27/Kip1 Ser10 phosphorylation and cytoplasmic relocalization. shRNA knockdown of p27 released the blockage of the mutant cells from cell cycle arrest at G 1 phase at a high-density condition. Together, these results indicate that vCyclin primarily functions to enhance cellular transformation and tumorigenesis by promoting cell cycle progression and cell proliferation at a contact-inhibited condition.


Assuntos
Carcinogênese/genética , Inibição de Contato/fisiologia , Ciclina D2/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Virais/metabolismo , Animais , Apoptose , Carcinogênese/patologia , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Senescência Celular , Ciclina D2/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Camundongos Nus , Mutação , Fosforilação , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA