Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Carcinogenesis ; 43(3): 277-287, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-34958370

RESUMEN

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.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Antiinflamatorios , Antioxidantes , Linfocitos B/metabolismo , Proliferación Celular , Humanos , Interleucina-6/metabolismo , Mitofagia , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/metabolismo
2.
Int J Cancer ; 147(12): 3500-3510, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32559816

RESUMEN

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.


Asunto(s)
Quimiocina CCL2/metabolismo , Células Endoteliales/citología , Herpesvirus Humano 8/patogenicidad , Mitocondrias/metabolismo , Sarcoma de Kaposi/virología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Endoteliales/virología , Transición Epitelial-Mesenquimal , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macroautofagia , Mitofagia , Modelos Biológicos , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Sarcoma de Kaposi/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Respuesta de Proteína Desplegada
3.
Br J Cancer ; 123(2): 298-306, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32418990

RESUMEN

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.


Asunto(s)
Antígeno B7-H1/genética , Endorribonucleasas/genética , Herpesvirus Humano 8/genética , Proteínas Serina-Treonina Quinasas/genética , Sarcoma de Kaposi/genética , Proteína 1 de Unión a la X-Box/genética , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Interleucina-10/genética , Interleucina-6/genética , Interleucina-8/genética , Activación de Macrófagos/genética , Macrófagos/virología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT6/genética , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/virología , Transducción de Señal , Activación Transcripcional/genética , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas Virales/genética , Replicación Viral/genética
4.
J Gen Virol ; 100(1): 89-98, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30427305

RESUMEN

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.


Asunto(s)
Autofagia , Estrés del Retículo Endoplásmico , Herpesvirus Humano 6/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Línea Celular , Genotipo , Herpesvirus Humano 6/genética , Humanos , Linfocitos T/patología , Linfocitos T/virología
5.
Int J Mol Sci ; 19(7)2018 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018188

RESUMEN

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.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias/genética , ARN Largo no Codificante/genética , ARN/genética , Regiones no Traducidas 3'/genética , Redes Reguladoras de Genes/genética , Humanos , Modelos Genéticos , Neoplasias/patología , Proteínas Proto-Oncogénicas/genética , ARN Circular
6.
Biochim Biophys Acta ; 1862(4): 805-813, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26769359

RESUMEN

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.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 2/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Especies Reactivas de Oxígeno/inmunología , Vía de Señalización Wnt/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Glucemia , Línea Celular , Células Dendríticas/patología , Diabetes Mellitus Tipo 2/patología , Humanos , Suero , beta Catenina/inmunología
7.
Biochim Biophys Acta ; 1853(7): 1586-95, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25827954

RESUMEN

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.


Asunto(s)
Autofagia , Herpesvirus Humano 4/fisiología , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Activación Viral , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antígenos Virales/metabolismo , Autofagia/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/virología , Ácido Butírico/farmacología , Supervivencia Celular/efectos de los fármacos , Herpesvirus Humano 4/efectos de los fármacos , Humanos , Modelos Biológicos , Fosforilación/efectos de los fármacos , Proteína Quinasa C-theta , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos
8.
Biochim Biophys Acta ; 1843(7): 1348-55, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24726834

RESUMEN

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.


Asunto(s)
Antígenos Virales/farmacología , Autofagia/efectos de los fármacos , Células Dendríticas/virología , Hepacivirus/inmunología , Sueros Inmunes/farmacología , Monocitos/virología , Inmunidad Adaptativa , Presentación de Antígeno , Antígenos CD1/genética , Antígenos CD1/inmunología , Antígenos Virales/inmunología , Autofagia/inmunología , Catepsina B/genética , Catepsina B/inmunología , Catepsina D/genética , Catepsina D/inmunología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/patología , Expresión Génica , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/patología , Hepatitis C Crónica/virología , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/inmunología , Monocitos/inmunología , Monocitos/patología
9.
Int J Cancer ; 137(6): 1491-7, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25704079

RESUMEN

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.


Asunto(s)
Linfocitos B/metabolismo , Diferenciación Celular/genética , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/genética , MicroARNs/genética , Animales , Sitios de Unión/genética , Ciclina D3/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación hacia Abajo/genética , Herpesvirus Humano 4 , Ratones , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/virología , Regiones Promotoras Genéticas
10.
J Virol ; 88(21): 12715-26, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25142602

RESUMEN

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.


Asunto(s)
Autofagia , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Evasión Inmune , Activación Viral , Replicación Viral , Animales , Línea Celular , Humanos
11.
Nucleic Acids Res ; 39(5): 1880-93, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21062812

RESUMEN

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.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/genética , Linfoma de Células B Grandes Difuso/virología , MicroARNs/metabolismo , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN/genética , Infecciones por Virus de Epstein-Barr/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Biblioteca de Genes , Herpesvirus Humano 4/metabolismo , Humanos , Linfoma de Células B Grandes Difuso/genética , MicroARNs/genética , Anotación de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo , ARN Pequeño no Traducido/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
12.
J Med Virol ; 84(5): 786-91, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22431027

RESUMEN

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.


Asunto(s)
Infecciones por Herpesviridae/transmisión , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiología , Saliva/virología , Sarcoma de Kaposi/virología , Secuencia de Aminoácidos , Línea Celular , ADN Viral/análisis , ADN Viral/sangre , ADN Viral/genética , Células Epiteliales/virología , Femenino , Genotipo , Células HEK293 , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/clasificación , Humanos , Italia/epidemiología , Leucocitos Mononucleares/virología , Masculino , Datos de Secuencia Molecular , Filogenia , Sarcoma de Kaposi/epidemiología , Análisis de Secuencia de ADN , Carga Viral , Tropismo Viral
13.
Virol J ; 9: 92, 2012 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-22583958

RESUMEN

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.


Asunto(s)
Movimiento Celular , Citoesqueleto/metabolismo , Células Dendríticas/inmunología , Herpesvirus Humano 8/inmunología , Receptores CCR6/biosíntesis , Receptores CCR7/biosíntesis , Células Dendríticas/virología , Regulación hacia Abajo , Herpesvirus Humano 8/patogenicidad , Humanos , Evasión Inmune
14.
Virus Res ; 292: 198231, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33207265

RESUMEN

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.


Asunto(s)
Antígeno B7-H1/genética , Citocinas/genética , Herpesvirus Humano 6/inmunología , Monocitos/microbiología , Especies Reactivas de Oxígeno/inmunología , Infecciones por Roseolovirus/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/inmunología , Antígeno B7-H1/inmunología , Citocinas/inmunología , Herpesvirus Humano 6/genética , Humanos , Monocitos/virología , Infecciones por Roseolovirus/genética , Infecciones por Roseolovirus/virología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/genética , Regulación hacia Arriba
15.
Microbes Infect ; 22(10): 585-591, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32882412

RESUMEN

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.


Asunto(s)
Autofagia , Herpesvirus Humano 4/fisiología , Proteínas de la Membrana/metabolismo , Proteínas Virales/metabolismo , Células HEK293 , Humanos , Lamina Tipo B/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Unión Proteica , Liberación del Virus , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
16.
Cells ; 9(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33297368

RESUMEN

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.


Asunto(s)
Autofagia , Herpesvirus Humano 4 , Herpesvirus Humano 6 , Herpesvirus Humano 8 , Virosis/patología , Infecciones por Herpesviridae/patología , Humanos , Sistema Inmunológico , Macrófagos/metabolismo , Neoplasias/metabolismo , Especies Reactivas de Oxígeno
17.
Biochim Biophys Acta Mol Basis Dis ; 1866(3): 165647, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31866416

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Astrocitoma/metabolismo , Autofagia/fisiología , Neuronas/metabolismo , Infecciones por Roseolovirus/metabolismo , Respuesta de Proteína Desplegada/fisiología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/virología , Astrocitoma/virología , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/fisiología , Herpesvirus Humano 6/patogenicidad , Humanos , Neuronas/virología , Fosforilación/fisiología , Infecciones por Roseolovirus/virología
18.
J Virol ; 82(8): 4042-51, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18287246

RESUMEN

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.


Asunto(s)
Herpesvirus Humano 4/fisiología , Proteínas Virales/fisiología , Ensamble de Virus , Línea Celular , Núcleo Celular/ultraestructura , Núcleo Celular/virología , Transformación Celular Viral/fisiología , Células Cultivadas , ADN Viral/biosíntesis , Eliminación de Gen , Prueba de Complementación Genética , Herpesvirus Humano 4/genética , Humanos , Leucocitos Mononucleares/virología , Microscopía Electrónica de Transmisión , Proteínas Virales/genética , Virión/ultraestructura
19.
Neural Regen Res ; 14(9): 1503-1506, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31089040

RESUMEN

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.
Virus Res ; 273: 197757, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31521763

RESUMEN

HHV-6A and HHV-6B are ubiquitous human betaherpesviruses sharing more than 80% homology. HHV-6B is the most common cause of encephalitis in transplant patients and its primary infection may cause the exanthema subitum and febrile seizures in infants. HHV-6A and HHV-6B are able to infect several immune cell types such as T cells, monocytes and dendritic cells (DCs). In this study we found that HHV-6 B derived from patients affected by exanthema subitum impaired monocyte differentiation into DCs, as the infected cells acquired less CD1a DC marker and retained more CD14 monocyte marker. In agreement with the previous finding that HHV-6B dysregulated autophagy and induced endoplasmic reticulum (ER) stress in cells in which it replicated, here we found that these effects occurred also in differentiating monocytes and that ER stress relief, by using the chemical chaperone sodium 4-phenylbutirate (PBA), partially restored DC formation. This suggests that the induction of ER stress, likely exacerbated by autophagy inhibition, could contribute to the immune suppression induced by HHV-6B derived from exanthema subitem patients.


Asunto(s)
Autofagia , Diferenciación Celular , Células Dendríticas/fisiología , Estrés del Retículo Endoplásmico , Herpesvirus Humano 6/fisiología , Monocitos/patología , Monocitos/virología , Supervivencia Celular , Células Cultivadas , Células Dendríticas/virología , Exantema Súbito/virología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA