Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Exp Med ; 220(1)2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36342455

RESUMEN

Inborn and acquired deficits of type I interferon (IFN) immunity predispose to life-threatening COVID-19 pneumonia. We longitudinally profiled the B cell response to mRNA vaccination in SARS-CoV-2 naive patients with inherited TLR7, IRF7, or IFNAR1 deficiency, as well as young patients with autoantibodies neutralizing type I IFNs due to autoimmune polyendocrine syndrome type-1 (APS-1) and older individuals with age-associated autoantibodies to type I IFNs. The receptor-binding domain spike protein (RBD)-specific memory B cell response in all patients was quantitatively and qualitatively similar to healthy donors. Sustained germinal center responses led to accumulation of somatic hypermutations in immunoglobulin heavy chain genes. The amplitude and duration of, and viral neutralization by, RBD-specific IgG serological response were also largely unaffected by TLR7, IRF7, or IFNAR1 deficiencies up to 7 mo after vaccination in all patients. These results suggest that induction of type I IFN is not required for efficient generation of a humoral response against SARS-CoV-2 by mRNA vaccines.


Asunto(s)
Linfocitos B , Vacunas contra la COVID-19 , COVID-19 , Interferón Tipo I , Humanos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Autoanticuerpos , COVID-19/inmunología , COVID-19/prevención & control , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética , Receptor Toll-Like 7/genética , Vacunación , Vacunas de ARNm , Vacunas contra la COVID-19/inmunología , Linfocitos B/inmunología , Interferón Tipo I/deficiencia
2.
J Virol ; 96(2): e0177421, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34757841

RESUMEN

Alphaviruses and flaviviruses have class II fusion glycoproteins that are essential for virion assembly and infectivity. Importantly, the tip of domain II is structurally conserved between the alphavirus and flavivirus fusion proteins, yet whether these structural similarities between virus families translate to functional similarities is unclear. Using in vivo evolution of Zika virus (ZIKV), we identified several novel emerging variants, including an envelope glycoprotein variant in ß-strand c (V114M) of domain II. We have previously shown that the analogous ß-strand c and the ij loop, located in the tip of domain II of the alphavirus E1 glycoprotein, are important for infectivity. This led us to hypothesize that flavivirus E ß-strand c also contributes to flavivirus infection. We generated this ZIKV glycoprotein variant and found that while it had little impact on infection in mosquitoes, it reduced replication in human cells and mice and increased virus sensitivity to ammonium chloride, as seen for alphaviruses. In light of these results and given our alphavirus ij loop studies, we mutated a conserved alanine at the tip of the flavivirus ij loop to valine to test its effect on ZIKV infectivity. Interestingly, this mutation inhibited infectious virion production of ZIKV and yellow fever virus, but not West Nile virus. Together, these studies show that shared domains of the alphavirus and flavivirus class II fusion glycoproteins harbor structurally analogous residues that are functionally important and contribute to virus infection in vivo.IMPORTANCE Arboviruses are a significant global public health threat, yet there are no antivirals targeting these viruses. This problem is in part due to our lack of knowledge of the molecular mechanisms involved in the arbovirus life cycle. In particular, virus entry and assembly are essential processes in the virus life cycle and steps that can be targeted for the development of antiviral therapies. Therefore, understanding common, fundamental mechanisms used by different arboviruses for entry and assembly is essential. In this study, we show that flavivirus and alphavirus residues located in structurally conserved and analogous regions of the class II fusion proteins contribute to common mechanisms of entry, dissemination, and infectious-virion production. These studies highlight how class II fusion proteins function and provide novel targets for development of antivirals.


Asunto(s)
Alphavirus/fisiología , Flavivirus/fisiología , Proteínas Virales de Fusión/metabolismo , Virión/metabolismo , Replicación Viral , Células A549 , Alphavirus/efectos de los fármacos , Cloruro de Amonio/farmacología , Animales , Culicidae/virología , Flavivirus/efectos de los fármacos , Humanos , Interferón Tipo I/deficiencia , Ratones , Ratones Mutantes , Mutación , Dominios Proteicos , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Virión/genética , Ensamble de Virus/genética , Internalización del Virus/efectos de los fármacos , Replicación Viral/genética , Virus Zika/efectos de los fármacos , Virus Zika/fisiología , Infección por el Virus Zika/virología
3.
Elife ; 102021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33416494

RESUMEN

Crimean-Congo hemorrhagic fever (CCHF) is a severe tick-borne febrile illness with wide geographic distribution. CCHF is caused by infection with the Crimean-Congo hemorrhagic fever virus (CCHFV) and case fatality rates can be as high as 30%. Despite causing severe disease in humans, our understanding of the host and viral determinants of CCHFV pathogenesis are limited. A major limitation in the investigation of CCHF has been the lack of suitable small animal models. Wild-type mice are resistant to clinical isolates of CCHFV and consequently, mice must be deficient in type I interferon responses to study the more severe aspects of CCHFV. We report here a mouse-adapted variant of CCHFV that recapitulates in adult, immunocompetent mice the severe CCHF observed in humans. This mouse-adapted variant of CCHFV significantly improves our ability to study host and viral determinants of CCHFV-induced disease in a highly tractable mouse model.


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo/fisiología , Fiebre Hemorrágica de Crimea/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Interferón Tipo I/deficiencia , Masculino , Ratones
4.
Nature ; 591(7849): 275-280, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33442058

RESUMEN

The innate immune regulator STING is a critical sensor of self- and pathogen-derived DNA. DNA sensing by STING leads to the induction of type-I interferons (IFN-I) and other cytokines, which promote immune-cell-mediated eradication of pathogens and neoplastic cells1,2. STING is also a robust driver of antitumour immunity, which has led to the development of STING activators and small-molecule agonists as adjuvants for cancer immunotherapy3. Pain, transmitted by peripheral nociceptive sensory neurons (nociceptors), also aids in host defence by alerting organisms to the presence of potentially damaging stimuli, including pathogens and cancer cells4,5. Here we demonstrate that STING is a critical regulator of nociception through IFN-I signalling in peripheral nociceptors. We show that mice lacking STING or IFN-I signalling exhibit hypersensitivity to nociceptive stimuli and heightened nociceptor excitability. Conversely, intrathecal activation of STING produces robust antinociception in mice and non-human primates. STING-mediated antinociception is governed by IFN-Is, which rapidly suppress excitability of mouse, monkey and human nociceptors. Our findings establish the STING-IFN-I signalling axis as a critical regulator of physiological nociception and a promising new target for treating chronic pain.


Asunto(s)
Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Nocicepción/fisiología , Dolor/metabolismo , Células Receptoras Sensoriales/metabolismo , Analgesia , Animales , Femenino , Humanos , Interferón Tipo I/deficiencia , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Macaca mulatta , Masculino , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Nocicepción/efectos de los fármacos , Transducción de Señal
5.
Mol Metab ; 34: 146-156, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32180554

RESUMEN

OBJECTIVE: In mouse models, deficiency of TTC39B (T39) decreases hepatic lipogenic gene expression and protects against diet-induced steatohepatitis. While assessing the therapeutic potential of antisense oligonucleotides (ASOs) targeting T39, we discovered an unexpected weight loss phenotype. The objective of this study was to determine the mechanism of the resistance to diet-induced obesity. METHODS: To assess therapeutic potential, we used antisense oligonucleotides (ASO) to knock down T39 expression in a Western or high-fat, high-cholesterol, high-sucrose-diet-fed Ldlr-/- or wild-type mice. RESULTS: T39 ASO treatment led to decreased hepatic lipogenic gene expression and decreased hepatic triglycerides. Unexpectedly, T39 ASO treatment protected against diet-induced obesity. The reduced weight gain was seen with two different ASOs that decreased T39 mRNA in adipose tissue macrophages (ATMs), but not with a liver-targeted GalNac-ASO. Mice treated with the T39 ASO displayed increased browning of gonadal white adipose tissue (gWAT) and evidence of increased lipolysis. However, T39 knockout mice displayed a similar weight loss response when treated with T39 ASO, indicating an off-target effect. RNA-seq analysis of gWAT showed a widespread increase in type I interferon (IFN)-responsive genes, and knockout of the IFN receptor abolished the weight loss phenotype induced by the T39 ASO. Some human T39 ASOs and ASOs with different modifications targeting LDLR also induced a type I IFN response in THP1 macrophages. CONCLUSION: Our data suggest that extrahepatic targeting of T39 by ASOs in ATMs produced an off-target type 1 IFN response, leading to activation of lipolysis, browning of WAT, and weight loss. While our findings suggest that ASOs may induce off-target type 1 IFN response more commonly than previously thought, they also suggest that therapeutic induction of type 1 IFN selectively in ATMs could potentially represent a novel approach to the treatment of obesity.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Interferón Tipo I/biosíntesis , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico , Oligonucleótidos Antisentido/farmacología , Animales , Femenino , Inyecciones Subcutáneas , Interferón Tipo I/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Obesidad/prevención & control , Oligonucleótidos Antisentido/administración & dosificación
6.
Antiviral Res ; 174: 104703, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932041

RESUMEN

For more than 20 years, researchers have used laboratory mice lacking type I or both type I and type II interferon (IFN) responses to study viruses that cause hemorrhagic fever (HF) in humans. Whereas immunocompetent mice do not become ill when infected with Ebola, Lassa, dengue and other HF viruses, IFN-deficient mice typically develop severe or fatal disease when inoculated with these pathogens. The ease of employment of these "mouse models" has led to their extensive use in biocontainment laboratories to assess the efficacy of novel vaccines, often without consideration of whether adaptive immune responses in IFN-deficient mice accurately mirror those in humans. Failure to consider these questions may lead to inappropriate expectations of the predictive value of mouse experiments. In two invited articles, we investigate this question. This paper examines how the lack of type I or both type I and type II IFN signaling may affect the development of adaptive immune responses in mice and the outcome of vaccine studies. A second article reviews the published literature on the use of IFN-deficient mice for the assessment of novel vaccines against HF viruses.


Asunto(s)
Inmunidad Adaptativa , Fiebres Hemorrágicas Virales/prevención & control , Interferón Tipo I/deficiencia , Interferón gamma/deficiencia , Vacunas Virales/inmunología , Animales , Modelos Animales de Enfermedad , Fiebres Hemorrágicas Virales/inmunología , Ratones , Vacunación
7.
Antiviral Res ; 174: 104702, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31982149

RESUMEN

For more than 20 years, researchers have used laboratory mice lacking type I or both type I and II interferon (IFN) responses to study high-containment viruses that cause hemorrhagic fevers (HF) in humans. With the exception of Rift Valley fever virus, agents that cause viral HF in humans, such as Ebola and Lassa virus, do not cause disease in mature immunocompetent mice. In contrast, IFN-deficient mice typically develop severe or fatal disease when inoculated with these agents. The sensitivity of IFN-deficient mice to disease has led to their widespread use in biocontainment laboratories to assess the efficacy of novel vaccines against HF viruses, often without considering whether adaptive immune responses in IFN-deficient mice accurately mirror those in immunocompetent humans. Failure to recognize these questions may lead to inappropriate expectations of the predictive value of mouse experiments. In two invited articles, we investigate these questions. The present article reviews the use of IFN-deficient mice for assessing novel vaccines against HF viruses, including Ebola, Lassa, Crimean-Congo hemorrhagic fever and Rift Valley fever viruses. A companion paper examines the general question of how the lack of IFN signaling may affect adaptive immune responses and the outcome of vaccine studies in mice.


Asunto(s)
Modelos Animales de Enfermedad , Fiebres Hemorrágicas Virales/inmunología , Fiebres Hemorrágicas Virales/prevención & control , Interferón Tipo I/deficiencia , Interferón gamma/deficiencia , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Ratones
8.
Curr Opin Pediatr ; 31(6): 815-820, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31693592

RESUMEN

PURPOSE OF REVIEW: Studying primary immunodeficiencies (PIDs) provides insights into human antiviral immunity in the natural infectious environment. This review describes new PIDs with genetic defects that impair innate antiviral responses. RECENT FINDINGS: New genetic defects in the interferon (IFN) signaling pathway include IFNAR1 deficiency, which causes uncontrolled infections with measles-mumps-rubella or yellow fever vaccines, and possibly also cytomegalovirus (CMV); and IRF9 deficiency, which results in influenza virus susceptibility. Genetic defects in several pattern recognition receptors include MDA5 deficiency, which impairs viral RNA sensing and confers human rhinovirus susceptibility; RNA polymerase III haploinsufficiency, which impairs sensing of A:T-rich virus DNA and confers VZV susceptibility; and TLR3 deficiency, which causes HSV-1 encephalitis (HSE) or influenza virus pneumonitis. Defects in RNA metabolism, such as that caused by Debranching enzyme 1 deficiency, can cause virus meningoencephalitis. Finally, defects in host restriction factors for virus replication, such as in CIB1 deficiency, contribute to uncontrolled ß-HPV infections. SUMMARY: Several new PIDs highlight the role of type I/III IFN signaling pathway, virus sensors, and host virus restriction factors in human antiviral immunity.


Asunto(s)
Antivirales/uso terapéutico , Inmunidad Innata/efectos de los fármacos , Gripe Humana , Interferón Tipo I/genética , Enfermedades de Inmunodeficiencia Primaria/tratamiento farmacológico , Humanos , Interferón Tipo I/deficiencia , Transducción de Señal
9.
Antiviral Res ; 168: 28-35, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31078648

RESUMEN

Influenza infections remain a burden on health care systems despite vaccination programs and marketed antiviral drugs. Immunomodulation through activation of innate sensors could represent innovative approaches to fight the flu. This study evaluated the ability of flagellin, agonist of Toll-like receptor 5 (TLR5), to control the replication of influenza A virus (IAV) in mice. First, we showed that systemic or intranasal administration of flagellin activated transcription of anti-viral genes in lung tissue. Prophylactic and therapeutic flagellin administration resulted in decreased levels of viral RNA and infectious virus in the lungs of H3N2 IAV-infected mice. The effect of the flagellin on viral replication was also observed in Ifnar-/- and Il22-/- IAV-infected mice, suggesting a mechanism independent of type I interferon and interleukin 22 signaling. In addition, a combination therapy associating the neuraminidase inhibitor oseltamivir and flagellin was more effective than standalone treatments in reducing pulmonary viral replication. Thus, this study highlights the therapeutic potential of the flagellin to control the replication of the influenza virus.


Asunto(s)
Antivirales/administración & dosificación , Flagelina/administración & dosificación , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oseltamivir/administración & dosificación , Receptor Toll-Like 5/agonistas , Replicación Viral/efectos de los fármacos , Animales , Virus de la Influenza A/efectos de los fármacos , Interferón Tipo I/deficiencia , Interleucinas/deficiencia , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Interleucina-22
10.
mBio ; 9(4)2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018108

RESUMEN

Gammaherpesviruses are oncogenic pathogens that persist in ~95% of the adult population. Cellular metabolic pathways have emerged as important regulators of many viral infections, including infections by gammaherpesviruses that require several lipid synthetic pathways for optimal replication. Liver X receptors (LXRs) are transcription factors that are critical regulators of cellular fatty acid and cholesterol synthesis pathways. Not surprisingly, LXRs are attractive therapeutic targets in cardiovascular disease. Here we describe an antiviral role for LXRs in the context of gammaherpesvirus infection of primary macrophages. We show that type I interferon increased LXR expression following infection. Surprisingly, there was not a corresponding induction of LXR target genes. Rather, LXRs suppressed the expression of target genes, leading to decreased fatty acid and cholesterol synthesis, two metabolic pathways that support gammaherpesvirus replication. This report defines LXR-mediated restriction of cholesterol and lipid synthesis as an intrinsic metabolic mechanism to restrict viral replication in innate immune cells.IMPORTANCE Fatty acid and cholesterol synthesis pathways of the host play important roles in diverse biological systems. Importantly, these two metabolic pathways are also usurped by a number of viruses to facilitate viral replication. In this report, we show that suppression of these pathways by liver X receptors in primary macrophages creates an intrinsic antiviral state that attenuates gammaherpesvirus replication by limiting viral access to the two metabolic pathways.


Asunto(s)
Antivirales/metabolismo , Colesterol/metabolismo , Ácidos Grasos/metabolismo , Gammaherpesvirinae/fisiología , Interacciones Huésped-Patógeno , Receptores X del Hígado/metabolismo , Replicación Viral , Animales , Antivirales/agonistas , Vías Biosintéticas/genética , Células Cultivadas , ADN Viral/metabolismo , Expresión Génica/efectos de los fármacos , Interferón Tipo I/deficiencia , Interferón Tipo I/farmacología , Receptores X del Hígado/agonistas , Receptores X del Hígado/deficiencia , Receptores X del Hígado/genética , Macrófagos/metabolismo , Macrófagos/virología , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo
11.
PLoS Negl Trop Dis ; 12(6): e0006524, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29927940

RESUMEN

Zika virus (ZIKV) has emerged since 2013 as a significant global human health threat following outbreaks in the Pacific Islands and rapid spread throughout South and Central America. Severe congenital and neurological sequelae have been linked to ZIKV infections. Assessing the ability of common mosquito species to transmit ZIKV and characterizing variation in mosquito transmission of different ZIKV strains is important for estimating regional outbreak potential and for prioritizing local mosquito control strategies for Aedes and Culex species. In this study, we evaluated the laboratory vector competence of Aedes aegypti, Culex quinquefasciatus, and Culex tarsalis that originated in areas of California where ZIKV cases in travelers since 2015 were frequent. We compared infection, dissemination, and transmission rates by measuring ZIKV RNA levels in cohorts of mosquitoes that ingested blood meals from type I interferon-deficient mice infected with either a Puerto Rican ZIKV strain from 2015 (PR15), a Brazilian ZIKV strain from 2015 (BR15), or an ancestral Asian-lineage Malaysian ZIKV strain from 1966 (MA66). With PR15, Cx. quinquefasciatus was refractory to infection (0%, N = 42) and Cx. tarsalis was infected at 4% (N = 46). No ZIKV RNA was detected in saliva from either Culex species 14 or 21 days post feeding (dpf). In contrast, Ae. aegypti developed infection rates of 85% (PR15; N = 46), 90% (BR15; N = 20), and 81% (MA66; N = 85) 14 or 15 dpf. Although MA66-infected Ae. aegypti showed higher levels of ZIKV RNA in mosquito bodies and legs, transmission rates were not significantly different across virus strains (P = 0.13, Fisher's exact test). To confirm infectivity and measure the transmitted ZIKV dose, we enumerated infectious ZIKV in Ae. aegypti saliva using Vero cell plaque assays. The expectorated plaque forming units PFU varied by viral strain: MA66-infected expectorated 13±4 PFU (mean±SE, N = 13) compared to 29±6 PFU for PR15-infected (N = 13) and 35±8 PFU for BR15-infected (N = 6; ANOVA, df = 2, F = 3.8, P = 0.035). These laboratory vector competence results support an emerging consensus that Cx. tarsalis and Cx. quinquefasciatus are not vectors of ZIKV. These results also indicate that Ae. aegypti from California are efficient laboratory vectors of ancestral and contemporary Asian lineage ZIKV.


Asunto(s)
Aedes/virología , Culex/virología , Mosquitos Vectores/virología , ARN Viral/aislamiento & purificación , Infección por el Virus Zika/transmisión , Virus Zika/fisiología , Animales , California/epidemiología , Chlorocebus aethiops , Humanos , Interferón Tipo I/deficiencia , Interferón Tipo I/inmunología , Ratones , Control de Mosquitos , Saliva/virología , Células Vero , Virus Zika/genética , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/virología
12.
J Immunol ; 198(11): 4210-4215, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28468974

RESUMEN

Interleukin-18 and IL-1ß, which are cytokines of the IL-1 family, are synthesized as precursor proteins and activated by the inflammasome via proteolytic processing. IL-1ß is only induced in response to inflammatory stimuli, but IL-18 is constitutively expressed. However, how IL-18 and IL-1ß expression is regulated by different inflammatory signals remains poorly studied. In this study, we found that IL-18 and IL-1ß are differentially regulated. Despite being constitutively expressed, IL-18 expression was increased and sustained after stimulation of TLRs. In contrast, IL-1ß was induced but not sustained after chronic treatment. Furthermore, type I IFN signaling was essential for induction of IL-18 and macrophages lacking type I IFN signaling were impaired in their ability to promote IL-18 induction. Thus, our findings reveal a fundamental difference in IL-18 and IL-1ß regulation and uncover novel mechanisms that are relevant to the inflammatory settings where these proinflammatory cytokines play a critical role.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-18/genética , Interleucina-1beta/genética , Transducción de Señal , Aminoquinolinas/farmacología , Animales , Células Cultivadas , Imidazoles/farmacología , Inflamasomas/inmunología , Interferón Tipo I/deficiencia , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interleucina-18/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Transducción de Señal/genética , Receptores Toll-Like/inmunología
13.
Arterioscler Thromb Vasc Biol ; 36(2): 266-73, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26634654

RESUMEN

OBJECTIVE: Patients with systemic lupus erythematosis are at risk for premature atherosclerosis and half of the patients with systemic lupus erythematosis have elevated type I interferon (IFN-I) levels. We hypothesized that IFN-I would induce premature atherosclerosis by increasing the number of smooth muscle progenitor cells (SMPC) in the bloodstream and promoting atherosclerotic lesions within the vasculature. APPROACH AND RESULTS: SMPC isolated from wild-type and IFN receptor knockout animals were cultured in medium±IFN-I. In vivo, we used electroporation to generate stable IFN-I expression for as long as 4 months. The number of SMPC was determined in mice that expressed IFN-I and in control mice and sections from the bifurcation of the abdominal aorta were analyzed 3 months after electroporation of an IFN-I expression plasmid or a control plasmid. Adding IFN-I to the media increased the number of cultured wild-type SMPC and increased mRNA for SM22, but had no effect on SMPC isolated from IFN receptor knockout mice. Our in vivo results demonstrated a positive relationship between the preatherosclerotic-like lesions and endothelial damage. Although, there were no significant differences in smooth muscle cell density or thickness of the medial layer between groups, the IFN-I-expressing mice had a significant increase in preatherosclerotic-like lesions and immature smooth muscle cells, cells that expressed CD34 and smooth muscle α-actin; but lacked smooth muscle myosin heavy chain. CONCLUSIONS: IFN-I seems to enhance SMPC number in vitro. In vivo IFN-I expression may maintain SMPC in an immature state. These immature smooth muscle cells could give rise to macrophages and eventually foam cells.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Diferenciación Celular , Interferón Tipo I/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Células Madre/metabolismo , Animales , Antígenos CD34/genética , Antígenos CD34/metabolismo , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Genotipo , Interferón Tipo I/deficiencia , Interferón Tipo I/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Cadenas Pesadas de Miosina/metabolismo , Fenotipo , Células Madre/patología , Factores de Tiempo , Transfección
14.
J Gen Virol ; 96(Pt 6): 1328-1339, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25646269

RESUMEN

The host and viral factors that influence disease outcome during flavivirus infections are not fully understood. Using the live attenuated yellow fever virus (YFV) vaccine strain 17D as a model system we evaluated how viral dose, inoculation route and immunopathogenesis contributed to disease outcome in mice deficient in the type I IFN response. We found that YFV-17D infection of IFN-α/ß receptor knockout mice resulted in three distinct disease outcomes: no clinical signs of disease, fatal viscerotropic disease or fatal neurotropic disease. Interestingly, viral load at disease onset did not correlate with disease outcome. However, we found increased immune infiltrates in the brain tissues of mice that developed neurotropic disease. Additionally, mice that developed viscerotropic disease, as characterized by liver and spleen pathology and/or intestinal haemorrhage, had significantly elevated levels of alanine aminotransferase, monocyte chemotactic protein and IFN-inducible protein (IP)-10 as compared with mice with no clinical signs of disease or neurotropic disease. Furthermore, mice treated with recombinant IP-10 throughout YFV-17D infection showed increased mortality and an increased percentage of mice with viscerotropic disease. Our results demonstrated that viral load did not correlate with pathogenesis, and the host immune response played a pivotal role in disease outcome and contributed to YFV-17D pathogenesis in mice.


Asunto(s)
Modelos Animales de Enfermedad , Fiebre Amarilla/patología , Fiebre Amarilla/virología , Virus de la Fiebre Amarilla/fisiología , Alanina Transaminasa/sangre , Animales , Encéfalo/patología , Quimiocina CCL2/sangre , Quimiocina CXCL10/sangre , Hemorragia Gastrointestinal , Interferón Tipo I/deficiencia , Intestinos/patología , Hígado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/patología , Análisis de Supervivencia , Carga Viral
15.
J Virol ; 89(2): 1479-83, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25392213

RESUMEN

The effect of abrogating the interferon (IFN) response on human cytomegalovirus (HCMV) replication was investigated using primary human cells engineered to block either the production of or the response to type I IFNs. In IFN-deficient cells, HCMV produced larger plaques and spread and replicated more rapidly than in parental cells. These cells demonstrate the vital role of IFNs in controlling HCMV replication and provide useful tools to investigate the IFN response to HCMV.


Asunto(s)
Citomegalovirus/inmunología , Citomegalovirus/fisiología , Interferón Tipo I/inmunología , Replicación Viral , Células Cultivadas , Humanos , Interferón Tipo I/deficiencia , Ensayo de Placa Viral
16.
J Neuropathol Exp Neurol ; 73(1): 59-71, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24335529

RESUMEN

The roles of Type I interferon (IFN) in human immunodeficiency virus Type 1 (HIV-1) neuropathogenesis are poorly understood; both protective and deleterious effects of IFN signaling have been described. We used genetically modified mice deficient in the Type I IFN receptor (IFNRKO) to analyze the progress of HIV-1 brain infection and neuropathogenesis in the absence of IFN signaling. IFNRKO and wild-type (WT) mice on the 129xSv/Ev or C57BL/6 strain backgrounds were infected systemically with EcoHIV, a chimeric HIV-1 that productively infects mice. IFNRKO mice showed higher HIV-1 expression in spleen and peritoneal macrophages and greater virus infiltration into the brain compared to WT mice. Neuropathogenesis was studied by histopathological, immunohistochemical, immunofluorescence, and polymerase chain reaction analyses of brain tissues after the virus was inoculated into the brain by stereotaxic intracerebral injection. Both IFNRKO and WT mice showed readily detectable HIV-1 and brain lesions, including microglial activation, astrocytosis, and increased expression of genes coding for inflammatory cytokines and chemokines typical of human HIV-1 brain disease. Parameters of HIV-1 neuropathogenesis, including HIV-1 expression in microglia/macrophages, were significantly greater in IFNRKO than in WT mice. Our results show unequivocally that Type I IFN signaling and responses limit HIV-1 infection and pathogenesis in the brains of mice.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , VIH-1/metabolismo , Interferón Tipo I/deficiencia , Animales , Regulación Viral de la Expresión Génica , Infecciones por VIH/genética , VIH-1/genética , Interferón Tipo I/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados
17.
J Heart Lung Transplant ; 32(9): 914-24, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23953819

RESUMEN

BACKGROUND: Respiratory RNA viruses are associated with bronchiolitis obliterans syndrome (BOS) in lung transplant recipients (LTRS); however, the immune mechanisms that regulate airway obliteration remain incompletely understood. METHODS: Using the mouse heterotopic tracheal transplant model of obliterative airway disease (OAD), we studied the role of double-stranded (ds)RNA using polyinosinic:polycytidylic acid (poly[I:C]), a synthetic analog of viral dsRNA, in abrogating airway allograft tolerance established with donor-specific transfusion (DST) and anti-CD154 monoclonal antibody therapy. RESULTS: Wild-type (WT) B6 recipients of accepted BALB/c airway grafts demonstrated significantly reduced intragraft CD8+ T cells, with markedly impaired allospecific interferon (INF)-γ and tumor necrosis factor-α secretion, uncoupled from an activated phenotype, and evidence of proliferation. Administration of poly(I:C) to DST/anti-CD154-treated recipients restored OAD pathology and CD8+ alloeffector responses to levels observed in untreated mice. However, B6 type I IFN receptor-deficient (IFN-αßR(-/-)) recipients were resistant to the abrogation of tolerance mediated by poly(I:C) and did not develop CD8+ alloeffector responses or OAD. Further, adoptive transfers of WT CD8+ T cells or CD11c+ dendritic cells alone into B6 IFNαßR(-/-) recipients treated with poly(I:C) and DST/anti-CD154 were incapable of abrogating airway graft tolerance. CONCLUSIONS: Together, these data indicate abrogation of DST/anti-CD154-induced airway allograft tolerance via dsRNA requires type-I IFN responsiveness for mouse airway obliteration.


Asunto(s)
Bronquiolitis Obliterante/cirugía , Rechazo de Injerto/inducido químicamente , Rechazo de Injerto/prevención & control , Inductores de Interferón/efectos adversos , Interferón Tipo I/fisiología , Poli I-C/efectos adversos , Tráquea/trasplante , Aloinjertos , Animales , Anticuerpos Monoclonales/uso terapéutico , Bronquiolitis Obliterante/fisiopatología , Ligando de CD40/inmunología , Linfocitos T CD8-positivos/patología , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto/fisiopatología , Interferón Tipo I/deficiencia , Interferón Tipo I/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , ARN Bicatenario/efectos adversos , Resultado del Tratamiento
18.
J Immunol ; 189(5): 2488-501, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22851707

RESUMEN

Localized elevation in type I IFN has been uniquely linked to the severe Lyme arthritis that develops in C3H mice infected with the spirochete Borrelia burgdorferi. In this study, the dynamic interactions that result in generation of these responses were further examined in C3H mice carrying the type I IFN receptor gene ablation, which effectively blocks all autocrine/paracrine signaling crucial to induction of downstream effectors. Reciprocal radiation chimeras between C3H and IFNAR1⁻/⁻ mice implicated both radiation-sensitive and radiation-resistant cells of the joint tissue in the proarthritic induction of type I IFN. Ex vivo analysis of cells from the naive joint revealed CD45⁺ cells residing in the tissue to be uniquely capable of initiating the type I IFN response to B. burgdorferi. Type I IFN responses were analyzed in real time by lineage sorting of cells from infected joint tissue. This demonstrated that myeloid cells, endothelial cells, and fibroblasts were responsible for propagating the robust IFN response, which peaked at day 7 postinfection and rapidly resolved. Endothelial cells and fibroblasts were the dominant sources of IFN signature transcripts in the joint tissue. Fibroblasts were also the major early source of chemokines associated with polymorphonuclear leukocyte and monocyte/macrophage infiltration, thus providing a focal point for arthritis development. These findings suggest joint-localized interactions among related and unrelated stromal, endothelial, and myeloid cell lineages that may be broadly applicable to understanding the pathogeneses of diseases associated with type I IFN signature, including systemic lupus erythematosus and some rheumatoid arthritides.


Asunto(s)
Artritis Experimental/inmunología , Fibroblastos/inmunología , Interferón Tipo I/biosíntesis , Enfermedad de Lyme/inmunología , Células Mieloides/inmunología , Regulación hacia Arriba/inmunología , Animales , Articulación del Tobillo/inmunología , Articulación del Tobillo/microbiología , Articulación del Tobillo/patología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Borrelia burgdorferi/inmunología , Borrelia burgdorferi/patogenicidad , Células Cultivadas , Quimiocinas/biosíntesis , Quimiocinas/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Interferón Tipo I/deficiencia , Interferón Tipo I/genética , Enfermedad de Lyme/metabolismo , Enfermedad de Lyme/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Células Mieloides/patología , Índice de Severidad de la Enfermedad , Transducción de Señal/inmunología , Transcripción Genética/inmunología , Regulación hacia Arriba/genética
19.
Vaccine ; 30(6): 1154-61, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22178730

RESUMEN

Type I IFN and IL-12 are well documented to serve as so called "signal 3" cytokines, capable of facilitating CD8(+) T cell proliferation, effector function and memory formation. While their ability to serve in this capacity is well established, to date, no non-cytokine signal 3 mediators have been clearly identified. We have established a vaccine model system in which the primary CD8(+) T cell response is independent of either IL-12 or type I IFN receptors, but dependent on CD27/CD70 interactions. We show here that primary and secondary CD8(+) T cell responses are generated in the combined deficiency of IFN and IL-12 signaling. In contrast, antigen specific CD8(+) T cell responses are compromised in the absence of the TNF receptors CD27 and OX40. These data indicate that CD27/OX40 can serve the central function as signal 3 mediators, independent of IFN or IL-12, for the generation of CD8(+) T cell immune memory.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Interferón Tipo I/inmunología , Interleucina-12/inmunología , Receptores OX40/metabolismo , Transducción de Señal , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Femenino , Interferón Tipo I/deficiencia , Interleucina-12/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
20.
PLoS Pathog ; 7(11): e1002358, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22072972

RESUMEN

Viral diseases of the respiratory tract, which include influenza pandemic, children acute bronchiolitis, and viral pneumonia of the elderly, represent major health problems. Plasmacytoid dendritic cells play an important role in anti-viral immunity, and these cells were recently shown to express ChemR23, the receptor for the chemoattractant protein chemerin, which is expressed by epithelial cells in the lung. Our aim was to determine the role played by the chemerin/ChemR23 system in the physiopathology of viral pneumonia, using the pneumonia virus of mice (PVM) as a model. Wild-type and ChemR23 knock-out mice were infected by PVM and followed for functional and inflammatory parameters. ChemR23(-/-) mice displayed higher mortality/morbidity, alteration of lung function, delayed viral clearance and increased neutrophilic infiltration. We demonstrated in these mice a lower recruitment of plasmacytoid dendritic cells and a reduction in type I interferon production. The role of plasmacytoid dendritic cells was further addressed by performing depletion and adoptive transfer experiments as well as by the generation of chimeric mice, demonstrating two opposite effects of the chemerin/ChemR23 system. First, the ChemR23-dependent recruitment of plasmacytoid dendritic cells contributes to adaptive immune responses and viral clearance, but also enhances the inflammatory response. Second, increased morbidity/mortality in ChemR23(-/-) mice is not due to defective plasmacytoid dendritic cells recruitment, but rather to the loss of an anti-inflammatory pathway involving ChemR23 expressed by non-leukocytic cells. The chemerin/ChemR23 system plays important roles in the physiopathology of viral pneumonia, and might therefore be considered as a therapeutic target for anti-viral and anti-inflammatory therapies.


Asunto(s)
Factores Quimiotácticos/metabolismo , Células Dendríticas/inmunología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Virus de la Neumonía Murina/inmunología , Neumonía Viral/inmunología , Infecciones por Pneumovirus/inmunología , Receptores Acoplados a Proteínas G/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Quimiocinas , Factores Quimiotácticos/biosíntesis , Células Dendríticas/metabolismo , Mediadores de Inflamación , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Interferón Tipo I/biosíntesis , Interferón Tipo I/deficiencia , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de la Neumonía Murina/metabolismo , Virus de la Neumonía Murina/patogenicidad , Neumonía Viral/metabolismo , Infecciones por Pneumovirus/metabolismo , Receptores de Quimiocina , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...