Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Carcinogenesis ; 43(3): 277-287, 2022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-34958370

RESUMO

Reactive oxygen species (ROS) and DNA repair, respectively, promote and limit oncogenic transformation of B cells driven by Epstein-Barr virus (EBV). We have previously shown that EBV infection reduced autophagy in primary B lymphocytes and enhanced ROS and interleukin 6 (IL-6) release, promoting B-cell proliferation and immortalization. In this study, we explored the role of p62/SQSTM1, accumulated as a consequence of autophagy reduction in EBV-infected B lymphocytes, and found that it exerted a growth-suppressive effect in these cells. At the molecular level, we found that p62 counteracted IL-6 production and ROS increase by interacting with NRF2 and promoting mitophagy. Moreover, p62/NRF2 axis sustained the expression level of H2AX and ataxia-telangiectasia mutated (ATM), whose activation has been shown to have growth-suppressive effects during the first steps of EBV infection, before latency is established. In conclusion, this study shows for the first time that the accumulation of p62 and the activation of p62/axis counteracted EBV-driven proliferation of primary B lymphocytes.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Anti-Inflamatórios , Antioxidantes , Linfócitos B/metabolismo , Proliferação de Células , Humanos , Interleucina-6/metabolismo , Mitofagia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo
2.
Int J Cancer ; 147(12): 3500-3510, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-32559816

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of KS, an aggressive neoplasm that mainly occurs in immune-compromised patients. Spindle cells represent the main feature of this aggressive malignancy and arise from KSHV-infected endothelial cells undergoing endothelial to mesenchymal transition (EndMT), which changes their cytoskeletal composition and organization. As in epithelial to mesenchymal transition (EMT), EndMT is driven by transcription factors such as SNAI1 and ZEB1 and implies a cellular reprogramming mechanism regulated by several molecular pathways, particularly PI3K/AKT/MTOR. Here we found that KSHV activated MTOR and its targets 4EBP1 and ULK1 and reduced bulk macroautophagy and mitophagy to promote EndMT, activate ER stress/unfolded protein response (UPR), and increase the release of the pro-angiogenic and pro-inflammatory chemokine CCL2 by HUVEC cells. Our study suggests that the manipulation of macroautophagy, mitophagy and UPR and the interplay between the three could be a promising strategy to counteract EndMT, angiogenesis and inflammation, the key events of KSHV-driven sarcomagenesis.


Assuntos
Quimiocina CCL2/metabolismo , Células Endoteliais/citologia , Herpesvirus Humano 8/patogenicidade , Mitocôndrias/metabolismo , Sarcoma de Kaposi/virologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/virologia , Transição Epitelial-Mesenquimal , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macroautofagia , Mitofagia , Modelos Biológicos , Cultura Primária de Células , Espécies Reativas de Oxigênio/metabolismo , Sarcoma de Kaposi/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Resposta a Proteínas não Dobradas
3.
Br J Cancer ; 123(2): 298-306, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32418990

RESUMO

BACKGROUND: Kaposi's Sarcoma Herpesvirus (KSHV) is a gammaherpesvirus strongly linked to human cancer. The virus is also able to induce immune suppression, effect that contributes to onset/progression of the viral-associated malignancies. As KSHV may infect macrophages and these cells abundantly infiltrate Kaposi's sarcoma lesions, in this study we investigated whether KSHV-infection could affect macrophage polarisation to promote tumorigenesis. METHODS: FACS analysis was used to detect macrophage markers and PD-L1 expression. KSHV infection and the molecular pathways activated were investigated by western blot analysis and by qRT-PCR while cytokine release was assessed by Multi-analyte Kit. RESULTS: We found that KSHV infection reduced macrophage survival and skewed their polarisation towards M2 like/TAM cells, based on the expression of CD163, on the activation of STAT3 and STAT6 pathways and the release of pro-tumorigenic cytokines such as IL-10, VEGF, IL-6 and IL-8. We also found that KSHV triggered Ire1 α-XBP1 axis activation in infected macrophages to increase the release of pro-tumorigenic cytokines and to up-regulate PD-L1 surface expression. CONCLUSIONS: The findings that KSHV infection of macrophages skews their polarisation towards M2/TAM and that activate Ire1 α-XBP1 to increase the release of pro-tumorigenic cytokines and the expression of PD-L1, suggest that manipulation of UPR could be exploited to prevent or improve the treatment of KSHV-associated malignancies.


Assuntos
Antígeno B7-H1/genética , Endorribonucleases/genética , Herpesvirus Humano 8/genética , Proteínas Serina-Treonina Quinases/genética , Sarcoma de Kaposi/genética , Proteína 1 de Ligação a X-Box/genética , Carcinogênese/genética , Regulação Neoplásica da Expressão Gênica/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Interleucina-10/genética , Interleucina-6/genética , Interleucina-8/genética , Ativação de Macrófagos/genética , Macrófagos/virologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT6/genética , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Transdução de Sinais , Ativação Transcricional/genética , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas Virais/genética , Replicação Viral/genética
4.
J Gen Virol ; 100(1): 89-98, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30427305

RESUMO

Herpesviruses are known to manipulate autophagy to optimize their replication, counteract immune response and probably to promote tumourigenesis. This study explored, for the first time, the impact of human herpesvirus (HHV)-6 lytic infection on autophagy and demonstrated that HHV-6A and B (viruses sharing more than 80 % homology) differently affected this cellular process. Indeed, while HHV-6A (GS) infection of HSB2 cells promoted autophagy, HHV-6B (Z29) or the virus isolated from the serum of roseola infantum-affected patient-inhibited autophagy in Molt-3 cells or in PBMCs, respectively. Interestingly, the different behaviour of HHV-6A and B on the autophagic process was accompanied by different effects on endoplasmic reticulum stress, unfolded protein response and cell survival that was more strongly reduced by HHV-6B infection. We hypothesize that the ability to inhibit autophagy displayed by HHV-6B could be due to the fact that it contains gene homologues of those encoding for TRS1; the protein responsible for the block of autophagy by human cytomegalovirus. Understanding how HHV-6A/B infection regulates autophagy could be of particular interest, as it has been recently shown that this virus may be involved in Alzheimer's disease in which a dysregulation of autophagy may also play a role.


Assuntos
Autofagia , Estresse do Retículo Endoplasmático , Herpesvirus Humano 6/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Linhagem Celular , Genótipo , Herpesvirus Humano 6/genética , Humanos , Linfócitos T/patologia , Linfócitos T/virologia
5.
Int J Mol Sci ; 19(7)2018 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-30018188

RESUMO

The past decade has witnessed enormous progress, and has seen the noncoding RNAs (ncRNAs) turn from the so-called dark matter RNA to critical functional molecules, influencing most physiological processes in development and disease contexts. Many ncRNAs interact with each other and are part of networks that influence the cell transcriptome and proteome and consequently the outcome of biological processes. The regulatory circuits controlled by ncRNAs have become increasingly more relevant in cancer. Further understanding of these complex network interactions and how ncRNAs are regulated, is paving the way for the identification of better therapeutic strategies in cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias/genética , RNA Longo não Codificante/genética , RNA/genética , Regiões 3' não Traduzidas/genética , Redes Reguladoras de Genes/genética , Humanos , Modelos Genéticos , Neoplasias/patologia , Proteínas Proto-Oncogênicas/genética , RNA Circular
6.
Biochim Biophys Acta ; 1862(4): 805-813, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26769359

RESUMO

Type 2 is the type of diabetes with higher prevalence in contemporary time, representing about 90% of the global cases of diabetes. In the course of diabetes, several complications can occur, mostly due to hyperglycemia and increased reactive oxygen species (ROS) production. One of them is represented by an increased susceptibility to microbial infections and by a reduced capacity to clear them. Therefore, knowing the impact of hyperglycemia on immune system functionality is of utmost importance for the management of the disease. In this study, we show that medium containing high glucose reduced the in-vitro differentiation of monocytes into functional DCs and their activation mediated by PAMPs or DAMPs. Most importantly, the same effects were mediated by the hyperglycemic sera derived by type 2 diabetic patients, mimicking a more physiologic condition. DC dysfunction caused by hyperglycemia may be involved in the inefficient control of infections observed in diabetic patients, given the pivotal role of these cells in both the innate and adaptive immune response. Searching for the molecular mechanisms underlying DC dysfunction, we found that canonical Wnt/ß-catenin and p38 MAPK pathways were activated in the DCs differentiated either in the presence of high glucose or of hyper-glycemic sera. Interestingly, the activation of these pathways and the DC immune dysfunction were partially counteracted by the anti-oxidant quercetin, a flavonoid already known to exert several beneficial effects in diabetes.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 2/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Espécies Reativas de Oxigênio/imunologia , Via de Sinalização Wnt/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Glicemia , Linhagem Celular , Células Dendríticas/patologia , Diabetes Mellitus Tipo 2/patologia , Humanos , Soro , beta Catenina/imunologia
7.
Biochim Biophys Acta ; 1853(7): 1586-95, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25827954

RESUMO

PKC activation by combining TPA with sodium butyrate (T/B) represents the most effective and widely used strategy to induce the Epstein-Barr virus (EBV) lytic cycle. The results obtained in this study show that novel PKCθ is involved in such process and that it acts through the activation of p38 MAPK and autophagy induction. Autophagy, a mechanism of cellular defense in stressful conditions, is manipulated by EBV to enhance viral replication. Besides promoting the EBV lytic cycle, the activation of p38 and autophagy resulted in a pro-survival effect, as indicated by p38 or ATG5 knocking down experiments. However, this pro-survival role was counteracted by a pro-death activity of PKCθ, due to the dephosphorylation of AKT. In conclusion, this study reports, for the first time, that T/B activates a PKCθ-p38 MAPK axis in EBV infected B cells, that promotes the viral lytic cycle and cell survival and dephosphorylates AKT, balancing cell life and cell death.


Assuntos
Autofagia , Herpesvirus Humano 4/fisiologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Ativação Viral , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Antígenos Virais/metabolismo , Autofagia/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/virologia , Ácido Butírico/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Herpesvirus Humano 4/efeitos dos fármacos , Humanos , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Proteína Quinase C-theta , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos
8.
Biochim Biophys Acta ; 1843(7): 1348-55, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24726834

RESUMO

Autophagy has a pivotal role in the in-vitro monocyte differentiation into macrophages and dendritic cells (DCs), the most powerful antigen presenting cells (APC) with the unique capacity to initiate an adaptive immune response. Autophagy is also a mechanism by which these cells of innate immunity may degrade intracellular pathogens and mediate the antigen processing and presentation, essential to clear an infection. For these reasons, pathogens have learned how to manipulate autophagy for their own survival. In this study we found that hepatitis C virus (HCV), derived from sera of infected patients, blocked the autophagic process in differentiating monocytes, seen as LC3 II and p62 expression levels. The suppression of autophagy correlated with a reduction of cathepsins D, B and proteolytic activity, and resulted in impairment of monocyte differentiation into DCs, as indicated by the reduction of CD1a acquirement. These data suggest that the block of autophagy might be one of the underlying mechanisms of the HCV-mediated immune subversion that frequently leads to viral persistence and chronic hepatitis.


Assuntos
Antígenos Virais/farmacologia , Autofagia/efeitos dos fármacos , Células Dendríticas/virologia , Hepacivirus/imunologia , Soros Imunes/farmacologia , Monócitos/virologia , Imunidade Adaptativa , Apresentação de Antígeno , Antígenos CD1/genética , Antígenos CD1/imunologia , Antígenos Virais/imunologia , Autofagia/imunologia , Catepsina B/genética , Catepsina B/imunologia , Catepsina D/genética , Catepsina D/imunologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/patologia , Expressão Gênica , Hepatite C Crônica/imunologia , Hepatite C Crônica/patologia , Hepatite C Crônica/virologia , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/imunologia , Monócitos/imunologia , Monócitos/patologia
9.
Int J Cancer ; 137(6): 1491-7, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25704079

RESUMO

The association of Epstein-Barr virus (EBV) with plasmacytoid malignancies is now well established but how the virus influences microRNA expression in such cells is not known. We have used multiple myeloma (MM) cell lines to address this issue and find that an oncomiR, miR-21 is induced after in vitro EBV infection. The PU.1 binding site in miR-21 promoter was essential for its activation by the virus. In accordance with its noted oncogenic functions, miR-21 induction in EBV infected MM cells caused downregulation of p21 and an increase in cyclin D3 expression. EBV infected MM cells were highly tumorigenic in SCID mice. Given the importance of miR-21 in plasmacytoid malignancies, our findings that EBV could further exacerbate the disease by inducing miR-21 has interesting implications both in terms of diagnosis and future miR based therapeutical approaches for the virus associated plasmacytoid tumors.


Assuntos
Linfócitos B/metabolismo , Diferenciação Celular/genética , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/genética , MicroRNAs/genética , Animais , Sítios de Ligação/genética , Ciclina D3/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação para Baixo/genética , Herpesvirus Humano 4 , Camundongos , Camundongos SCID , Mieloma Múltiplo/genética , Mieloma Múltiplo/virologia , Regiões Promotoras Genéticas
10.
J Virol ; 88(21): 12715-26, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25142602

RESUMO

UNLABELLED: Autophagy is a catabolic pathway that helps cells to survive under stressful conditions. Cells also use autophagy to clear microbiological infections, but microbes have learned how to manipulate the autophagic pathway for their own benefit. The experimental evidence obtained in this study suggests that the autophagic flux is blocked at the final steps during the reactivation of Epstein-Barr virus (EBV) from latency. This is indicated by the level of the lipidated form of LC3 that does not increase in the presence of bafilomycin and by the lack of colocalization of autophagosomes with lysosomes, which correlates with reduced Rab7 expression. Since the inhibition of the early phases of autophagy impaired EBV replication and viral particles were observed in autophagic vesicles in the cytoplasm of producing cells, we suggest that EBV exploits the autophagic machinery for its transportation in order to enhance viral production. The autophagic block was not mediated by ZEBRA, an immediate-early EBV lytic gene, whose transfection in Ramos, Akata, and 293 cells promoted a complete autophagic flux. The block occurred only when the complete set of EBV lytic genes was expressed. We suggest that the inhibition of the early autophagic steps or finding strategies to overcome the autophagic block, allowing viral degradation into the lysosomes, can be exploited to manipulate EBV replication. IMPORTANCE: This study shows, for the first time, that autophagy is blocked at the final degradative steps during EBV replication in several cell types. Through this block, EBV hijacks the autophagic vesicles for its intracellular transportation and enhances viral production. A better understanding of virus-host interactions could help in the design of new therapeutic approaches against EBV-associated malignancies.


Assuntos
Autofagia , Herpesvirus Humano 4/fisiologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Ativação Viral , Replicação Viral , Animais , Linhagem Celular , Humanos
11.
Nucleic Acids Res ; 39(5): 1880-93, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21062812

RESUMO

The Epstein-Barr virus (EBV) is an oncogenic human Herpes virus found in ∼15% of diffuse large B-cell lymphoma (DLBCL). EBV encodes miRNAs and induces changes in the cellular miRNA profile of infected cells. MiRNAs are small, non-coding RNAs of ∼19-26 nt which suppress protein synthesis by inducing translational arrest or mRNA degradation. Here, we report a comprehensive miRNA-profiling study and show that hsa-miR-424, -223, -199a-3p, -199a-5p, -27b, -378, -26b, -23a, -23b were upregulated and hsa-miR-155, -20b, -221, -151-3p, -222, -29b/c, -106a were downregulated more than 2-fold due to EBV-infection of DLBCL. All known EBV miRNAs with the exception of the BHRF1 cluster as well as EBV-miR-BART15 and -20 were present. A computational analysis indicated potential targets such as c-MYB, LATS2, c-SKI and SIAH1. We show that c-MYB is targeted by miR-155 and miR-424, that the tumor suppressor SIAH1 is targeted by miR-424, and that c-SKI is potentially regulated by miR-155. Downregulation of SIAH1 protein in DLBCL was demonstrated by immunohistochemistry. The inhibition of SIAH1 is in line with the notion that EBV impedes various pro-apoptotic pathways during tumorigenesis. The down-modulation of the oncogenic c-MYB protein, although counter-intuitive, might be explained by its tight regulation in developmental processes.


Assuntos
Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/genética , Linfoma Difuso de Grandes Células B/virologia , MicroRNAs/metabolismo , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA/genética , Infecções por Vírus Epstein-Barr/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Biblioteca Gênica , Herpesvirus Humano 4/metabolismo , Humanos , Linfoma Difuso de Grandes Células B/genética , MicroRNAs/genética , Anotação de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-myb/genética , Proteínas Proto-Oncogênicas c-myb/metabolismo , Pequeno RNA não Traduzido/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
12.
J Med Virol ; 84(5): 786-91, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22431027

RESUMO

The study was performed to determine if there is an association between the genotype and transmission of HHV-8 types A and C. These HHV-8 subtypes are prevalent in the area of North of Sardinia, which is an island off west Italy's mainland that has a high HHV-8 seroprevalence (35%). Blood and saliva samples from 30 patients with classic Kaposi's sarcoma who were lifetime residents of North Sardinia were analyzed to identify the HHV-8 genotype and quantitate the viral load. Genotype A, especially A1 subtype, was found more frequently (9/30 patients) and had a significantly higher viral load in saliva compared to blood (P = 0.029), where type C was found more frequently but with a viral load lower than 10(3) copies/ml. To determine if there is a correlation between the viral genotype and cellular tropism, type A1 and C3 HHV-8 viral particles were obtained from cell lines BCBL1 and BC3 infected chronically with HHV-8 A1 and C3 genotypes respectively and used to infect HEK293 epithelial origin cells and PBMCs in vitro. The data indicate that the A1 HHV-8 genotype is tropic and replicates at higher levels in the epithelial cell lines.


Assuntos
Infecções por Herpesviridae/transmissão , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Saliva/virologia , Sarcoma de Kaposi/virologia , Sequência de Aminoácidos , Linhagem Celular , DNA Viral/análise , DNA Viral/sangue , DNA Viral/genética , Células Epiteliais/virologia , Feminino , Genótipo , Células HEK293 , Infecções por Herpesviridae/epidemiologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/classificação , Humanos , Itália/epidemiologia , Leucócitos Mononucleares/virologia , Masculino , Dados de Sequência Molecular , Filogenia , Sarcoma de Kaposi/epidemiologia , Análise de Sequência de DNA , Carga Viral , Tropismo Viral
13.
Virol J ; 9: 92, 2012 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-22583958

RESUMO

BACKGROUND: For an efficient immune response against viral infection, dendritic cells (DCs) must express a coordinate repertoire of receptors that allow their recruitment to the sites of inflammation and subsequently to the secondary lymphoid organs in response to chemokine gradients.Several pathogens are able to subvert the chemokine receptor expression and alter the migration properties of DCs as strategy to escape from the immune control. FINDINGS: Here we report the inhibitory effect of Human Herpesvirus 8 (HHV-8) on the migratory behavior of immature and mature DCs. We found that the virus altered the DC chemokine receptor expression and chemokine induced migration. Moreover HHV-8 was also able to interfere with basal motility of DCs by inducing cytoskeleton modifications. CONCLUSION: Based on our findings, we suggest that HHV-8 is able to subvert the DC migration capacity and this represents an additional mechanism which interferes with their immune-functions.


Assuntos
Movimento Celular , Citoesqueleto/metabolismo , Células Dendríticas/imunologia , Herpesvirus Humano 8/imunologia , Receptores CCR6/biossíntese , Receptores CCR7/biossíntese , Células Dendríticas/virologia , Regulação para Baixo , Herpesvirus Humano 8/patogenicidade , Humanos , Evasão da Resposta Imune
14.
Virus Res ; 292: 198231, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33207265

RESUMO

Programmed death ligand 1 (PD-L1) up-regulation on antigen presenting cells induces T cell dysfunction, strongly impairing immune response. Human Herpesviruses (HHV) 6B is a ß-herpesvirus that, although displays a higher tropism for T cells, can infect other immune cells including monocytes and dendritic cells (DCs) and neuronal cells. We have previously shown that HHV-6B infection of primary monocytes reduced autophagy and induced Endoplasmic Reticulum (ER) stress/ Unfolded Protein Response (UPR), impairing their survival and differentiation into DCs. In this study, we found that PD-L1 expression was up-regulated by HHV-6B on the surface of infected monocytes and that its extracellular release also increased, effects known to lead to an impairment of anti-viral immune response. At molecular level, PD-L1 up-regulation correlated with the activation of a positive regulatory circuit between the increase of intracellular ROS and the activation of STAT1 and STAT3 induced by HHV-6B, accompanied by a high release of pro-inflammatory/immune suppressive cytokines. In conclusion, this study unveils new strategies put in place by HHV-6B to induce immune dysfunction and the underlying molecular pathways that could be targeted to counteract such immune suppressive effects.


Assuntos
Antígeno B7-H1/genética , Citocinas/genética , Herpesvirus Humano 6/imunologia , Monócitos/microbiologia , Espécies Reativas de Oxigênio/imunologia , Infecções por Roseolovirus/imunologia , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT3/imunologia , Antígeno B7-H1/imunologia , Citocinas/imunologia , Herpesvirus Humano 6/genética , Humanos , Monócitos/virologia , Infecções por Roseolovirus/genética , Infecções por Roseolovirus/virologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT3/genética , Regulação para Cima
15.
Biochim Biophys Acta Mol Basis Dis ; 1866(3): 165647, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31866416

RESUMO

HHV-6A and HHV-6B are neurotropic viruses able to dysregulate autophagy and activate ER stress/UPR in several cell types. The appropriate functioning of these processes is required for cell homeostasis, particularly in post-mitotic cells such as neuronal cells. Interestingly, neurodegenerative diseases such as Alzheimer's disease (AD) are often accompanied by autophagy dysregulation and abnormal UPR activation. This study demonstrated for the first time that HHV-6A infection of astrocytoma cells and primary neurons reduces autophagy, increases Aß production and activates ER stress/UPR promoting tau protein hyper-phosphorylation. Our results support previous studies suggesting that HHV-6A infection may play a role in AD and unveil the possible underlying molecular mechanisms involved.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Astrocitoma/metabolismo , Autofagia/fisiologia , Neurônios/metabolismo , Infecções por Roseolovirus/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Proteínas tau/metabolismo , Doença de Alzheimer/virologia , Astrocitoma/virologia , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/fisiologia , Herpesvirus Humano 6/patogenicidade , Humanos , Neurônios/virologia , Fosforilação/fisiologia , Infecções por Roseolovirus/virologia
16.
Cells ; 9(12)2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33297368

RESUMO

Human Herpes Virus-6 (HHV-6), Epstein-Barr Virus (EBV) and Kaposi Sarcoma Herpes Virus (KSHV) are viruses that share with other member of the Herpesvirus family the capacity to interfere with the autophagic process. In this paper, mainly based on the findings of our laboratory, we describe how, through different mechanisms, these viruses converge in reducing autophagy to impair DC immune function and how, by infecting and dysregulating autophagy in different cell types, they promote the pathologies associated with their infection, from the neurodegenerative diseases such Alzheimer's disease to cancer.


Assuntos
Autofagia , Herpesvirus Humano 4 , Herpesvirus Humano 6 , Herpesvirus Humano 8 , Viroses/patologia , Infecções por Herpesviridae/patologia , Humanos , Sistema Imunitário , Macrófagos/metabolismo , Neoplasias/metabolismo , Espécies Reativas de Oxigênio
17.
Microbes Infect ; 22(10): 585-591, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32882412

RESUMO

Viral egress and autophagy are two mechanisms that seem to be strictly connected in Herpesviruses's biology. Several data suggest that the autophagic machinery facilitates the egress of viral capsids and thus the production of new infectious particles. In the Herpesvirus family, viral nuclear egress is controlled and organized by a well conserved group of proteins named Nuclear Egress Complex (NEC). In the case of EBV, NEC is composed by BFRF1 and BFLF2 proteins, although the alterations of the nuclear host cell architecture are mainly driven by BFRF1, a multifunctional viral protein anchored to the inner nuclear membrane of the host cell. BFRF1 shares a peculiar distribution with several nuclear components and with them it strictly interacts. In this study, we investigated the possible role of BFRF1 in manipulating autophagy, pathway that possibly originates from nucleus, regulating the interplay between autophagy and viral egress.


Assuntos
Autofagia , Herpesvirus Humano 4/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Virais/metabolismo , Células HEK293 , Humanos , Lamina Tipo B/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Ligação Proteica , Liberação de Vírus , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
18.
J Virol ; 82(8): 4042-51, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18287246

RESUMO

Previous genetic and biochemical studies performed with several members of the Alphaherpesvirus subfamily have shown that the UL31 and UL34 proteins are essential components of the molecular machinery that mediates the primary egress of newly assembled capsids across the nuclear membrane. Further, there is substantial evidence that BFLF2 and BFRF1, the respective positional homologs of UL31 and UL34 in the Epstein-Barr virus (EBV) genome, are also their functional homologs, i.e., that the UL31/UL34 pathway is common to distant herpesviruses. However, the low degree of protein sequence identity between UL31 and BFLF2 would argue against such a hypothesis. To further clarify this issue, we have constructed a recombinant EBV strain devoid of BFLF2 (DeltaBFLF2) and show that BFLF2 is crucial for efficient virus production but not for lytic DNA replication or B-cell transformation. This defective phenotype could be efficiently restored by trans complementation with a BFLF2 expression plasmid. Detailed analysis of replicating cells by electron microscopy revealed that, as expected, DeltaBFLF2 viruses not only failed to egress from the nucleus but also showed defective DNA packaging. Nonfunctional primary egress did not, however, impair the production and extracellular release of enveloped but empty viral particles that comprised L particles containing tegument-like structures and a few virus-like particles carrying empty capsids. The DeltaBFLF2 and DeltaUL31 phenotypes therefore only partly overlap, from which we infer that BFLF2 and UL31 have substantially diverged during evolution to fulfil related but distinct functions.


Assuntos
Herpesvirus Humano 4/fisiologia , Proteínas Virais/fisiologia , Montagem de Vírus , Linhagem Celular , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Transformação Celular Viral/fisiologia , Células Cultivadas , DNA Viral/biossíntese , Deleção de Genes , Teste de Complementação Genética , Herpesvirus Humano 4/genética , Humanos , Leucócitos Mononucleares/virologia , Microscopia Eletrônica de Transmissão , Proteínas Virais/genética , Vírion/ultraestrutura
19.
Neural Regen Res ; 14(9): 1503-1506, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31089040

RESUMO

Neurotropic herpesviruses have been associated with the onset and progression of Alzheimer's disease, a common form of dementia that afflicts a large percentage of elderly individuals. Interestingly, among the neurotropic herpesviruses, herpes simplex virus-1, human herpesvirus-6A, and human herpesvirus-6B have been reported to infect several cell types present in the central nervous system and to dysregulate autophagy, a process required for homeostasis of cells, especially neurons. Indeed autophagosome accumulation, indicating an unbalance between autophagosome formation and autophagosome degradation, has been observed in neurons of Alzheimer's disease patients and may play a role in the intracellular and extracellular accumulation of amyloid ß and in the altered protein tau metabolism. Moreover, herpesvirus infection of central nervous system cells such as glia and microglia can increase the production of oxidant species through the alteration of mitochondrial dynamics and promote inflammation, another hallmark of Alzheimer's disease. This evidence suggests that it is worth further investigating the role of neurotropic herpesviruses, particularly human herpesvirus-6A/B, in the etiopathogenesis of Alzheimer's disease.

20.
Int J Biochem Cell Biol ; 114: 105560, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31220583

RESUMO

Kaposi Sarcoma Herpes Virus (KSHV) is an oncovirus belonging to the human gammaherpesvirus family, able to infect several immune cell types including B cells, dendritic cells (DCs) and monocytes. In this study, we found that KSHV infection of monocytes counteracted the Reactive Oxygen Species (ROS) increase induced by Macrophage Colony-Stimulating Factor (M-CSF), prevented c-Jun N-terminal kinase (JNK) and B-cell lymphoma-2 (Bcl-2) phosphorylation and inhibited autophagy, leading to an impairment of cell survival and differentiation into macrophages. We also show that, to further dysregulate immune response in monocytes, KSHV reduced the production of pro-inflammatory cytokines such as Tumor necrosis factor alpha (TNF α) while increased the release of the immune suppressive cytokine Interleukin-10 (IL-10). These results unveils new strategies put in place by KSHV to induce immune suppression and to persist into the infected host.


Assuntos
Autofagia , Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8/metabolismo , MAP Quinase Quinase 4/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sobrevivência Celular , Infecções por Herpesviridae/patologia , Humanos , Interleucina-10/metabolismo , Macrófagos/patologia , Macrófagos/virologia , Fosforilação , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA