Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Diabetologia ; 67(5): 908-927, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38409439

RESUMEN

AIMS/HYPOTHESIS: The proinflammatory cytokines IFN-α, IFN-γ, IL-1ß and TNF-α may contribute to innate and adaptive immune responses during insulitis in type 1 diabetes and therefore represent attractive therapeutic targets to protect beta cells. However, the specific role of each of these cytokines individually on pancreatic beta cells remains unknown. METHODS: We used deep RNA-seq analysis, followed by extensive confirmation experiments based on reverse transcription-quantitative PCR (RT-qPCR), western blot, histology and use of siRNAs, to characterise the response of human pancreatic beta cells to each cytokine individually and compared the signatures obtained with those present in islets of individuals affected by type 1 diabetes. RESULTS: IFN-α and IFN-γ had a greater impact on the beta cell transcriptome when compared with IL-1ß and TNF-α. The IFN-induced gene signatures have a strong correlation with those observed in beta cells from individuals with type 1 diabetes, and the level of expression of specific IFN-stimulated genes is positively correlated with proteins present in islets of these individuals, regulating beta cell responses to 'danger signals' such as viral infections. Zinc finger NFX1-type containing 1 (ZNFX1), a double-stranded RNA sensor, was identified as highly induced by IFNs and shown to play a key role in the antiviral response in beta cells. CONCLUSIONS/INTERPRETATION: These data suggest that IFN-α and IFN-γ are key cytokines at the islet level in human type 1 diabetes, contributing to the triggering and amplification of autoimmunity.


Asunto(s)
Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Humanos , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Interferones/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interferón gamma/metabolismo , Islotes Pancreáticos/metabolismo
3.
Sci Adv ; 8(37): eabn5732, 2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36103539

RESUMEN

IFNα is a key regulator of the dialogue between pancreatic ß cells and the immune system in early type 1 diabetes (T1D). IFNα up-regulates HLA class I expression in human ß cells, fostering autoantigen presentation to the immune system. We observed by bulk and single-cell RNA sequencing that exposure of human induced pluripotent-derived islet-like cells to IFNα induces expression of HLA class I and of other genes involved in antigen presentation, including the transcriptional activator NLRC5. We next evaluated the global role of NLRC5 in human insulin-producing EndoC-ßH1 and human islet cells by RNA sequencing and targeted gene/protein determination. NLRC5 regulates expression of HLA class I, antigen presentation-related genes, and chemokines. NLRC5 also mediates the effects of IFNα on alternative splicing, a generator of ß cell neoantigens, suggesting that it is a central player of the effects of IFNα on ß cells that contribute to trigger and amplify autoimmunity in T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Islotes Pancreáticos , Empalme Alternativo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Interferón-alfa/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Islotes Pancreáticos/metabolismo , Transcripción Genética
4.
Diabetes Obes Metab ; 22(10): 1827-1836, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32476252

RESUMEN

AIM: Type 1 diabetes (T1D) is a chronic autoimmune disease leading to progressive loss of pancreatic beta cells. Interferon (IFN)-α plays a critical role in the crosstalk between pancreatic beta cells and the immune system in early insulitis. In human beta cells IFNα signals through JAK1 and TYK2, leading to endoplasmic reticulum stress, inflammation and HLA class I overexpression. IFNα, acting synergistically with IL-1ß, induces apoptosis. Polymorphisms in TYK2 that decrease its activity are associated with protection against T1D, and we hypothesized that pharmacological inhibitors that specifically target TYK2 could protect human beta cells against the deleterious effects of IFNα. MATERIALS AND METHODS: Two TYK2 inhibitors provided by Nimbus Lakshmi were tested in human insulin-producing EndoC-ßH1 cells and human islets to evaluate their effect on IFNα signalling, beta-cell function and susceptibility to viral infection using RT-qPCR, western blot, immunofluorescence, ELISA and nuclear dyes. RESULTS: The two TYK2 inhibitors tested prevented IFNα-induced human beta-cell gene expression in a dose-dependent manner. They also protected human islets against IFNα + IL-1ß-induced apoptosis. Importantly, these inhibitors did not modify beta-cell function or their survival following infection with the potential diabetogenic coxsackieviruses CVB1 and CVB5. CONCLUSIONS: The two TYK2 inhibitors tested inhibit the IFNα signalling pathway in human beta cells, decreasing its pro-inflammatory and pro-apoptotic effects without sensitizing the cells to viral infection. The preclinical findings could pave the way for future clinical trials with TYK2 inhibitors for the prevention and treatment of type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Apoptosis , Citoprotección , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Estrés del Retículo Endoplásmico , Humanos , TYK2 Quinasa/genética
5.
J Innate Immun ; 11(4): 375-390, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30799417

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease characterized by islet inflammation and progressive pancreatic ß cell destruction. The disease is triggered by a combination of genetic and environmental factors, but the mechanisms leading to the triggering of early innate and late adaptive immunity and consequent progressive pancreatic ß cell death remain unclear. The insulin-producing ß cells are active secretory cells and are thus particularly sensitive to endoplasmic reticulum (ER) stress. ER stress plays an important role in the pathologic pathway leading to autoimmunity, islet inflammation, and ß cell death. We show here that group B coxsackievirus (CVB) infection, a putative causative factor for T1D, induces a partial ER stress in rat and human ß cells. The activation of the PERK/ATF4/CHOP branch is blunted while the IRE1α branch leads to increased spliced XBP1 expression and c-Jun N-terminal kinase (JNK) activation. Interestingly, JNK1 activation is essential for CVB amplification in both human and rat ß cells. Furthermore, a chemically induced ER stress preceding viral infection increases viral replication, in a process dependent on IRE1α activation. Our findings show that CVB tailors the unfolded protein response in ß cells to support their replication, preferentially triggering the pro-viral IRE1α/XBP1s/JNK1 pathway while blocking the pro-apoptotic PERK/ATF4/CHOP pathway.


Asunto(s)
Infecciones por Coxsackievirus/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Enterovirus Humano B/fisiología , Células Secretoras de Insulina/inmunología , Respuesta de Proteína Desplegada/inmunología , Animales , Línea Celular , Infecciones por Coxsackievirus/inmunología , Diabetes Mellitus Tipo 1/inmunología , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Humanos , Evasión Inmune , Células Secretoras de Insulina/virología , MAP Quinasa Quinasa 4/metabolismo , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Transducción de Señal , Replicación Viral , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/metabolismo
6.
EBioMedicine ; 36: 367-375, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30269996

RESUMEN

BACKGROUND: Antibodies targeting PD-1 and its ligand PDL1 are used in cancer immunotherapy but may lead to autoimmune diseases, including type 1 diabetes (T1D). It remains unclear whether PDL1 is expressed in pancreatic islets of people with T1D and how is it regulated. METHODS: The expression of PDL1, IRF1, insulin and glucagon was evaluated in samples of T1D donors by immunofluorescence. Cytokine-induced PDL1 expression in the human beta cell line, EndoC-ßH1, and in primary human pancreatic islets was determined by real-time RT-PCR, flow cytometry and Western blot. Specific and previously validated small interference RNAs were used to inhibit STAT1, STAT2, IRF1 and JAK1 signaling. Key results were validated using the JAK inhibitor Ruxolitinib. FINDINGS: PDL1 was present in insulin-positive cells from twelve T1D individuals (6 living and 6 deceased donors) but absent from insulin-deficient islets or from the islets of six non-diabetic controls. Interferons-α and -γ, but not interleukin-1ß, induced PDL1 expression in vitro in human islet cells and EndoC-ßH1 cells. Silencing of STAT1 or STAT2 individually did not prevent interferon-α-induced PDL1, while blocking of JAKs - a proposed therapeutic strategy for T1D - or IRF1 prevented PDL1 induction. INTERPRETATION: These findings indicate that PDL1 is expressed in beta cells from people with T1D, possibly to attenuate the autoimmune assault, and that it is induced by both type I and II interferons via IRF1.


Asunto(s)
Antígeno B7-H1/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Regulación de la Expresión Génica , Factor 1 Regulador del Interferón/metabolismo , Interferón-alfa/metabolismo , Interferón gamma/metabolismo , Islotes Pancreáticos/metabolismo , Adolescente , Adulto , Biomarcadores , Línea Celular , Niño , Preescolar , Humanos , Células Secretoras de Insulina/metabolismo , Persona de Mediana Edad , Adulto Joven
7.
Trends Endocrinol Metab ; 29(3): 137-139, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29326001

RESUMEN

Enteroviruses are important environmental contributors to islet inflammation (insulitis) in type 1 diabetes mellitus (T1DM). A recent study characterized the proteomic alterations induced by Coxsackievirus type B (CVB) infection of human islets. This provides relevant information to decipher the words of the virus-induced 'dialog' between ß cells and the immune system that leads to autoimmunity.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Humanos , Proteómica
8.
Diabetologia ; 60(4): 656-667, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28062922

RESUMEN

AIMS/HYPOTHESIS: Three hallmarks of the pancreatic islets in early human type 1 diabetes are overexpression of HLA class I, endoplasmic reticulum (ER) stress and beta cell apoptosis. The mediators of these phenomena remain to be defined. The type I interferon IFNα is expressed in human islets from type 1 diabetes patients and mediates HLA class I overexpression. We presently evaluated the mechanisms involved in IFNα-induced HLA class I expression in human beta cells and determined whether this cytokine contributes to ER stress and apoptosis. METHODS: IFNα-induced inflammation, ER stress and apoptosis were evaluated by RT-PCR, western blot, immunofluorescence and nuclear dyes, and proteins involved in type I interferon signalling were inhibited by small interfering RNAs. All experiments were performed in human islets or human EndoC-ßH1 cells. RESULTS: IFNα upregulates HLA class I, inflammation and ER stress markers in human beta cells via activation of the candidate gene TYK2, and the transcription factors signal transducer and activator of transcription 2 and IFN regulatory factor 9. Furthermore, it acts synergistically with IL-1ß to induce beta cell apoptosis. CONCLUSIONS/INTERPRETATION: The innate immune effects induced by IFNα may induce and amplify the adaptive immune response against human beta cells, indicating that IFNα has a central role in the early phases of diabetes.


Asunto(s)
Apoptosis/fisiología , Diabetes Mellitus Tipo 1/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Secretoras de Insulina/metabolismo , Interferón-alfa/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 1/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Interleucina-1beta/farmacología , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Nat Rev Endocrinol ; 12(5): 263-273, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27020257

RESUMEN

Type 1 diabetes mellitus (T1DM) is caused by progressive autoimmune-mediated loss of pancreatic ß-cell mass via apoptosis. The onset of T1DM depends on environmental factors that interact with predisposing genes to induce an autoimmune assault against ß cells. Epidemiological, clinical and pathology studies in humans support viral infection--particularly by enteroviruses (for example, coxsackievirus)--as an environmental trigger for the development of T1DM. Many candidate genes for T1DM, such as MDA5, PTPN2 and TYK2, regulate antiviral responses in both ß cells and the immune system. Cellular permissiveness to viral infection is modulated by innate antiviral responses that vary among different tissues or cell types. Some data indicate that pancreatic islet α cells trigger a more efficient antiviral response to infection with diabetogenic viruses than do ß cells, and so are able to eradicate viral infections without undergoing apoptosis. This difference could account for the varying ability of islet-cell subtypes to clear viral infections and explain why chronically infected pancreatic ß cells, but not α cells, are targeted by an autoimmune response and killed during the development of T1DM. These issues and attempts to target viral infection as a preventive therapy for T1DM are discussed in the present Review.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/patología , Virosis/complicaciones , Virosis/patología , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/prevención & control , Humanos , Virosis/genética
10.
Diabetes ; 64(11): 3808-17, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26239055

RESUMEN

Pancreatic ß-cells are destroyed by an autoimmune attack in type 1 diabetes. Linkage and genome-wide association studies point to >50 loci that are associated with the disease in the human genome. Pathway analysis of candidate genes expressed in human islets identified a central role for interferon (IFN)-regulated pathways and tyrosine kinase 2 (TYK2). Polymorphisms in the TYK2 gene predicted to decrease function are associated with a decreased risk of developing type 1 diabetes. We presently evaluated whether TYK2 plays a role in human pancreatic ß-cell apoptosis and production of proinflammatory mediators. TYK2-silenced human ß-cells exposed to polyinosinic-polycitidilic acid (PIC) (a mimick of double-stranded RNA produced during viral infection) showed less type I IFN pathway activation and lower production of IFNα and CXCL10. These cells also had decreased expression of major histocompatibility complex (MHC) class I proteins, a hallmark of early ß-cell inflammation in type 1 diabetes. Importantly, TYK2 inhibition prevented PIC-induced ß-cell apoptosis via the mitochondrial pathway of cell death. The present findings suggest that TYK2 regulates apoptotic and proinflammatory pathways in pancreatic ß-cells via modulation of IFNα signaling, subsequent increase in MHC class I protein, and modulation of chemokines such as CXCL10 that are important for recruitment of T cells to the islets.


Asunto(s)
Apoptosis/genética , Diabetes Mellitus Tipo 1/genética , Inmunidad Innata/genética , Células Secretoras de Insulina/metabolismo , TYK2 Quinasa/genética , Apoptosis/inmunología , Línea Celular , Supervivencia Celular/genética , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Genes MHC Clase I/fisiología , Estudio de Asociación del Genoma Completo , Humanos , Células Secretoras de Insulina/inmunología , Interferón-alfa/genética , Interferón-alfa/metabolismo , Fosforilación , Polimorfismo de Nucleótido Simple , TYK2 Quinasa/metabolismo
11.
Elife ; 4: e06990, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26061776

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease caused by loss of pancreatic ß cells via apoptosis while neighboring α cells are preserved. Viral infections by coxsackieviruses (CVB) may contribute to trigger autoimmunity in T1D. Cellular permissiveness to viral infection is modulated by innate antiviral responses, which vary among different cell types. We presently describe that global gene expression is similar in cytokine-treated and virus-infected human islet cells, with up-regulation of gene networks involved in cell autonomous immune responses. Comparison between the responses of rat pancreatic α and ß cells to infection by CVB5 and 4 indicate that α cells trigger a more efficient antiviral response than ß cells, including higher basal and induced expression of STAT1-regulated genes, and are thus better able to clear viral infections than ß cells. These differences may explain why pancreatic ß cells, but not α cells, are targeted by an autoimmune response during T1D.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/virología , Células Secretoras de Glucagón/inmunología , Células Secretoras de Glucagón/virología , Inmunidad Innata , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/virología , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratas Wistar
12.
Vet J ; 196(3): 381-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23159676

RESUMEN

Parvoviruses depend on initiation of host cell division for their replication. Undefined parvoviral proteins have been detected in Purkinje cells of the cerebellum after experimental feline panleukopenia virus (FPV) infection of neonatal kittens and in naturally occurring cases of feline cerebellar hypoplasia. In this study, a parvoviral protein in the nucleus of Purkinje cells of kittens with cerebellar hypoplasia was shown by immunoprecipitation to be the FPV viral capsid protein VP2. In PCR-confirmed, FPV-associated feline cerebellar hypoplasia, expression of the FPV VP2 protein was demonstrated by immunohistochemistry in Purkinje cell nuclei in 4/10 cases and expression of the FPV non-structural protein NS1 was demonstrated in Purkinje cell nuclei in 5/10 cases. Increased nuclear ERK1 expression was observed in several Purkinje cells in 1/10 kittens. No expression of the G1 and S mitotic phase marker proliferating cell nuclear antigen (PCNA) was evident in Purkinje cell nuclei. These results support the hypothesis that FPV is able to proceed far into its replication cycle in post-mitotic Purkinje cells.


Asunto(s)
Enfermedades de los Gatos/virología , Cerebelo/anomalías , Virus de la Panleucopenia Felina/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Malformaciones del Sistema Nervioso/veterinaria , Células de Purkinje/virología , Proteínas Virales/metabolismo , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales , Especificidad de Anticuerpos , Enfermedades de los Gatos/patología , Gatos , Cerebelo/citología , Cerebelo/metabolismo , Cerebelo/virología , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/virología , Virus de la Panleucopenia Felina/genética , Inmunohistoquímica/veterinaria , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/virología , Proteínas Virales/genética
13.
Hepatology ; 55(4): 998-1007, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22121002

RESUMEN

UNLABELLED: Hepatitis C virus (HCV) particles are known to be in complex with lipoproteins. As a result of this interaction, the low-density lipoprotein (LDL) receptor (LDLR) has been proposed as a potential entry factor for HCV; however, its implication in virus entry remains unclear. Here, we reinvestigated the role of the LDLR in the HCV life cycle by comparing virus entry to the mechanism of lipoprotein uptake. A small interfering RNA targeting the LDLR in Huh-7 cells reduced HCV infectivity, confirming that this receptor plays a role in the life cycle of HCV generated in cell culture. However, kinetics of internalization were much faster for lipoproteins than for infectious HCV particles. Furthermore, a decrease in HCV RNA replication was observed by blocking the LDLR with a specific antibody, and this was associated with an increase in the ratio of phosphatidylethanolamine to phosphatidylcholine in host cells. Nevertheless, a soluble form of the LDLR inhibited both HCV entry into the hepatocytes and its binding to the LDLR expressed on Chinese hamster ovary cells, suggesting a direct interaction between the HCV particle and the LDLR. Finally, we showed that modification of HCV particles by lipoprotein lipase (LPL) reduces HCV infectivity and increases HCV binding to LDLR. Importantly, LPL treatment also induced an increase in RNA internalization, suggesting that LDLR, at least in some conditions, leads to nonproductive internalization of HCV. CONCLUSION: The LDLR is not essential for infectious HCV particle entry, whereas the physiological function of this receptor is important for optimal replication of the HCV genome.


Asunto(s)
Hepacivirus/crecimiento & desarrollo , Hepacivirus/fisiología , Estadios del Ciclo de Vida/fisiología , Receptores de LDL/fisiología , Animales , Anticuerpos/farmacología , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Femenino , Células HEK293 , Hepacivirus/patogenicidad , Hepatocitos/patología , Hepatocitos/virología , Humanos , Riñón/citología , Riñón/virología , Ovario/citología , Ovario/virología , ARN Viral/metabolismo , Receptores de LDL/antagonistas & inhibidores , Receptores de LDL/efectos de los fármacos , Replicación Viral/fisiología
14.
PLoS Pathog ; 7(9): e1002267, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21977009

RESUMEN

The rise in type 1 diabetes (T1D) incidence in recent decades is probably related to modifications in environmental factors. Viruses are among the putative environmental triggers of T1D. The mechanisms regulating beta cell responses to viruses, however, remain to be defined. We have presently clarified the signaling pathways leading to beta cell apoptosis following exposure to the viral mimetic double-stranded RNA (dsRNA) and a diabetogenic enterovirus (Coxsackievirus B5). Internal dsRNA induces cell death via the intrinsic mitochondrial pathway. In this process, activation of the dsRNA-dependent protein kinase (PKR) promotes eIF2α phosphorylation and protein synthesis inhibition, leading to downregulation of the antiapoptotic Bcl-2 protein myeloid cell leukemia sequence 1 (Mcl-1). Mcl-1 decrease results in the release of the BH3-only protein Bim, which activates the mitochondrial pathway of apoptosis. Indeed, Bim knockdown prevented both dsRNA- and Coxsackievirus B5-induced beta cell death, and counteracted the proapoptotic effects of Mcl-1 silencing. These observations indicate that the balance between Mcl-1 and Bim is a key factor regulating beta cell survival during diabetogenic viral infections.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Infecciones por Coxsackievirus/metabolismo , Enterovirus Humano B/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Animales , Proteína 11 Similar a Bcl2 , Línea Celular , Supervivencia Celular , Infecciones por Coxsackievirus/patología , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/virología , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/virología , Masculino , Mitocondrias/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Fosforilación , Ratas , Ratas Wistar
15.
Hepatology ; 50(1): 6-16, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19437494

RESUMEN

UNLABELLED: Statins are 3-hydroxyl-3-methylglutaryl coenzyme A (HMG CoA) reductase inhibitors used for the treatment of hypercholesterolemia. It was recently reported that statins inhibit in vitro hepatitis C virus (HCV) RNA replication. We here report that, of five statins studied, mevastatin and simvastatin exhibit the strongest in vitro anti-HCV activity, lovastatin and fluvastatin have moderate inhibitory effects, and pravastatin is devoid of an antiviral effect. A combination of statins with interferon-alpha (IFN-alpha) or HCV nonstructural (NS)5B polymerase or NS3 protease inhibitors results in an additive antiviral activity in short-term (3 days) antiviral assays. Neither statins, at a concentration of five-fold their median effective concentration (EC(50)) value, nor polymerase, protease inhibitors, or IFN-alpha, at concentrations 10- or 20-fold their EC(50) value, were able to clear cells from their replicon following four or six consecutive passages of antiviral pressure. However, the combination of HCV polymerase or protease inhibitors with mevastatin or simvastatin resulted in an efficient clearance of the cultures from their replicon. In colony formation experiments, mevastatin reduced the frequency or prevented the selection of HCV replicons resistant to the nonnucleoside inhibitor HCV-796. CONCLUSION: A combination of specific HCV inhibitors with statins may result in a more profound antiviral effect and may delay or prevent the development of resistance to such inhibitors.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Células Cultivadas , Sinergismo Farmacológico , Factores de Tiempo
16.
Cell Microbiol ; 10(3): 606-17, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17979982

RESUMEN

Virus entry is a major step in which host-cell lipids can play an essential role. In this report, we investigated the importance of sphingolipids in hepatitis C virus (HCV) entry. For this purpose, sphingomyelin present in the plasma membrane of target cells was hydrolysed into ceramide by sphingomyelinase treatment. Interestingly, ceramide enrichment of the plasma membrane strongly inhibited HCV entry. To understand how ceramide affected HCV entry, we analysed the effect of ceramide enrichment of the plasma membrane on three cell-surface molecules identified as entry factors for HCV: CD81 tetraspanin, scavenger receptor BI and Claudin-1. These proteins, which we found to be mainly associated with detergent-soluble membranes in Huh-7 cells, were not relocated in detergent-resistant microdomains after sphingomyelin hydrolysis into ceramide. Importantly, ceramide enrichment of the plasma membrane led to a 50% decrease in cell-surface CD81, which was due to its ATP-independent internalization. Our results strongly suggest that the ceramide-induced internalization of CD81 is responsible for the inhibitory effect of ceramide on HCV entry. Together, these data indicate that some specific lipids of the plasma membrane are essential for HCV entry and highlight plasma membrane lipids as potential targets to block HCV entry.


Asunto(s)
Antígenos CD/metabolismo , Membrana Celular/metabolismo , Ceramidas/metabolismo , Hepacivirus/fisiología , Internalización del Virus , Línea Celular , Claudina-1 , Humanos , Proteínas de la Membrana/metabolismo , Receptores Depuradores de Clase B/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/metabolismo , Tetraspanina 28
17.
Mol Cancer Ther ; 5(2): 391-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16505114

RESUMEN

Non-small cell lung cancers (NSCLC) are associated with very dismal prognoses, and adjuvant chemotherapy, including irinotecan, taxanes, platin, and Vinca alkaloid derivatives, offers patients only slight clinical benefits. Part of the chemoresistance of NSCLCs results from the constitutive or anticancer drug-induced activation of the nuclear factor-kappaB (NF-kappaB) signaling pathways. The present study shows that human A549 NSCLC cells display highly activated cytoprotective NF-kappaB signaling pathways. UNBS1450, which is a cardenolide belonging to the same class of chemicals as ouabain and digitoxin, affected the expression and activation status of different constituents of the NF-kappaB pathways in these A549 tumor cells. The modifications brought about by UNBS1450 led to a decrease in both the DNA-binding capacity of the p65 subunit and the NF-kappaB transcriptional activity. Using the 3-(4,5-dimethylthiazol-2yl)-dephenyltetrazolium bromide colorimetric assay, we observed in vitro that UNBS1450 was as potent as taxol and SN38 (the active metabolite of irinotecan) in reducing the overall growth levels of the human A549 NSCLC cell line, and was more efficient than platin derivatives, including cisplatin, carboplatin, and oxaliplatin. The chronic in vivo i.p. and p.o. UNBS1450 treatments of human A549 orthotopic xenografts metastasizing to the brains and the livers of immunodeficient mice had a number of significant therapeutic effects on this very aggressive model.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cardenólidos/farmacología , Citoprotección/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , FN-kappa B/metabolismo , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cardenólidos/uso terapéutico , ADN/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Factor de Transcripción ReIA/metabolismo , Transcripción Genética , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Biol Chem ; 280(9): 7793-9, 2005 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-15632171

RESUMEN

The scavenger receptor class B type I (SR-BI) has recently been shown to interact with hepatitis C virus (HCV) envelope glycoprotein E2, suggesting that it might be involved at some step of HCV entry into host cells. However, due to the absence of a cell culture system to efficiently amplify HCV, it is not clear how SR-BI contributes to HCV entry. Here, we sought to determine how high density lipoproteins (HDLs), the natural ligand of SR-BI, affect HCV entry. By using the recently described infectious HCV pseudotyped particles (HCVpps) that display functional E1E2 glycoprotein complexes, we showed that HDLs are able to markedly enhance HCVpp entry. We did not find any evidence of HDL association with HCVpps, suggesting that HCVpps do not enter into target cells using HDL as a carrier to bind to its receptor. Interestingly, lipid-free apoA-I and apoA-II, the major HDL apolipoproteins, were unable to enhance HCVpp infectivity. In addition, drugs inhibiting HDL cholesteryl transfer (block lipid transport (BLT)-2 and BLT-4) reduced HDL enhancement of HCVpp entry, suggesting a role for lipid transfer in facilitating HCVpp entry. Importantly, silencing of SR-BI expression in target cells by RNA interference markedly reduced HDL-mediated enhancement of HCVpp entry. Finally, enhancement of HCVpp entry was also suppressed when the SR-BI binding region on HCV glycoprotein E2 was deleted. Altogether, these data indicate that HDL-mediated enhancement of HCVpp entry involves a complex interplay between SR-BI, HDL, and HCV envelope glycoproteins, and they highlight the active role of HDLs in HCV entry.


Asunto(s)
Hepacivirus/metabolismo , Lipoproteínas HDL/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas del Envoltorio Viral/química , Antígenos CD36 , Línea Celular , Centrifugación por Gradiente de Densidad , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Silenciador del Gen , Glicoproteínas/química , Humanos , Inmunoprecipitación , Ligandos , Lípidos/química , Lipoproteínas HDL/química , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Depuradores , Receptores Depuradores de Clase B , Sacarosa/farmacología , Proteínas del Envoltorio Viral/metabolismo
19.
J Virol ; 78(22): 12591-602, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15507646

RESUMEN

The immature flavivirus particle contains two envelope proteins, prM and E, that are associated as a heterodimer. Virion morphogenesis of the flaviviruses occurs in association with endoplasmic reticulum (ER) membranes, suggesting that there should be accumulation of the virion components in this compartment. This also implies that ER localization signals must be present in the flavivirus envelope proteins. In this work, we looked for potential subcellular localization signals in the yellow fever virus envelope proteins. Confocal immunofluorescence analysis of the subcellular localization of the E protein in yellow fever virus-infected cells indicated that this protein accumulates in the ER. Similar results were obtained with cells expressing only prM and E. Chimeric proteins containing the ectodomain of CD4 or CD8 fused to the transmembrane domains of prM or E were constructed, and their subcellular localization was studied by confocal immunofluorescence and by analyzing the maturation of their associated glycans. Although a small fraction was detected in the ER-to-Golgi intermediate and Golgi compartments, these chimeric proteins were located mainly in the ER. The C termini of prM and E form two antiparallel transmembrane alpha-helices. Interestingly, the first transmembrane passage contains enough information for ER localization. Taken altogether, these data indicate that, besides their role as membrane anchors, the transmembrane domains of yellow fever virus envelope proteins are ER retention signals. In addition, our data show that the mechanisms of ER retention of the flavivirus and hepacivirus envelope proteins are different.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Virus de la Fiebre Amarilla/química , Secuencia de Aminoácidos , Antígenos CD4/análisis , Membrana Celular/química , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/análisis , Proteínas del Envoltorio Viral/química
20.
J Virol ; 78(17): 9224-32, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15308717

RESUMEN

Mechanisms of virion attachment, interaction with its receptor, and cell entry are poorly understood for hepatitis C virus (HCV) because of a lack of an efficient and reliable in vitro system for virus propagation. Infectious HCV retroviral pseudotype particles (HCVpp) were recently shown to express native E1E2 glycoproteins, as defined in part by HCV human monoclonal antibodies (HMAbs) to conformational epitopes on E2, and some of these antibodies block HCVpp infection (A. Op De Beeck, C. Voisset, B. Bartosch, Y. Ciczora, L. Cocquerel, Z. Y. Keck, S. Foung, F. L. Cosset, and J. Dubuisson, J. Virol. 78:2994-3002, 2004). Why some HMAbs are neutralizing and others are nonneutralizing is looked at in this report by a series of studies to determine the expression of their epitopes on E2 associated with HCVpp and the role of antibody binding affinity. Antibody cross-competition defined three E2 immunogenic domains with neutralizing HMAbs restricted to two domains that were also able to block E2 interaction with CD81, a putative receptor for HCV. HCVpp immunoprecipitation showed that neutralizing and nonneutralizing domains are expressed on E2 associated with HCVpp, and affinity studies found moderate-to-high-affinity antibodies in all domains. These findings support the perspective that HCV-specific epitopes are responsible for functional steps in virus infection, with specific antibodies blocking distinct steps of virus attachment and entry, rather than the perspective that virus neutralization correlates with increased antibody binding to any virion surface site, independent of the epitope recognized by the antibody. Segregation of virus neutralization and sensitivity to low pH to specific regions supports a model of HCV E2 immunogenic domains similar to the antigenic structural and functional domains of other flavivirus envelope E glycoproteins.


Asunto(s)
Epítopos/química , Epítopos/inmunología , Hepacivirus/fisiología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Afinidad de Anticuerpos , Unión Competitiva , Línea Celular , Reacciones Cruzadas , Células HeLa , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Hepacivirus/inmunología , Humanos , Concentración de Iones de Hidrógeno , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/inmunología , Pruebas de Neutralización , Estructura Terciaria de Proteína , Eliminación de Secuencia , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA