Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 8(5): e1002715, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615572

RESUMEN

Epstein-Barr virus (EBV) has been identified as a putative environmental trigger of multiple sclerosis (MS), yet EBV's role in MS remains elusive. We utilized murine gamma herpesvirus 68 (γHV-68), the murine homolog to EBV, to examine how infection by a virus like EBV could enhance CNS autoimmunity. Mice latently infected with γHV-68 developed more severe EAE including heightened paralysis and mortality. Similar to MS, γHV-68EAE mice developed lesions composed of CD4 and CD8 T cells, macrophages and loss of myelin in the brain and spinal cord. Further, T cells from the CNS of γHV-68 EAE mice were primarily Th1, producing heightened levels of IFN-γ and T-bet accompanied by IL-17 suppression, whereas a Th17 response was observed in uninfected EAE mice. Clearly, γHV-68 latency polarizes the adaptive immune response, directs a heightened CNS pathology following EAE induction reminiscent of human MS and portrays a novel mechanism by which EBV likely influences MS and other autoimmune diseases.


Asunto(s)
Encéfalo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/virología , Rhadinovirus/patogenicidad , Médula Espinal/inmunología , Latencia del Virus , Animales , Encéfalo/patología , Encéfalo/virología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/patología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Herpesvirus Humano 4/patogenicidad , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Médula Espinal/patología , Médula Espinal/virología , Proteínas de Dominio T Box/biosíntesis , Células TH1/inmunología , Células Th17/inmunología
2.
J Immunol ; 188(4): 1638-46, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22250093

RESUMEN

P-selectin glycoprotein ligand-1 (PSGL-1), a heavily glycosylated sialomucin expressed on most leukocytes, has dual function as a selectin ligand for leukocyte rolling on vascular selectins expressed in inflammation and as a facilitator of resting T cell homing into lymphoid organs. In this article, we document disturbances in T cell homeostasis present in PSGL-1(null) mice. Naive CD4(+) and CD8(+) T cell frequencies were profoundly reduced in blood, whereas T cell numbers in lymph nodes and spleen were at or near normal levels. Although PSGL-1(null) T cells were less efficient at entering lymph nodes, they also remained in lymph nodes longer than PSGL-1(+/+) T cells, suggesting that PSGL-1 supports T cell egress. In addition, PSGL-1(null) CD8(+) T cell proliferation was observed under steady-state conditions and PSGL-1(null) CD8(+) T cells were found to be hyperresponsive to homeostatic cytokines IL-2, IL-4, and IL-15. Despite these disturbances in T cell homeostasis, PSGL-1(null) mice exhibited a normal acute response (day 8) to lymphocytic choriomeningitis virus infection but generated an increased frequency of memory T cells (day 40). Our observations demonstrate a novel pleiotropic influence of PSGL-1 deficiency on several aspects of T cell homeostasis that would not have been anticipated based on the mild phenotype of PSGL-1(null) mice. These potentially offsetting effects presumably account for the near-normal cellularity seen in lymph nodes of PSGL-1(null) mice.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/fisiología , Movimiento Celular , Células Cultivadas , Homeostasis , Interleucina-1/inmunología , Interleucina-2/inmunología , Interleucina-4/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Virus de la Coriomeningitis Linfocítica/inmunología , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL
3.
Sci Rep ; 12(1): 21189, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36477199

RESUMEN

Age-associated B cells (ABCs; CD19+CD11c+T-bet+) are a unique population that are increased in an array of viral infections, though their role during latent infection is largely unexplored. Here, we use murine gammaherpesvirus 68 (γHV68) to demonstrate that ABCs remain elevated long-term during latent infection and express IFNγ and TNF. Using a recombinant γHV68 that is cleared following acute infection, we show that ABCs persist in the absence of latent virus, though their expression of IFNγ and TNF is decreased. With a fluorescent reporter gene-expressing γHV68 we demonstrate that ABCs are infected with γHV68 at similar rates to other previously activated B cells. We find that mice without ABCs display defects in anti-viral IgG2a/c antibodies and are more susceptible to reactivation of γHV68 following virus challenges that typically do not break latency. Together, these results indicate that ABCs are a persistent effector subset during latent viral infection that impedes γHV68 reactivation.


Asunto(s)
Infección Latente , Animales , Ratones
4.
Front Immunol ; 13: 858583, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35874728

RESUMEN

Epstein-Barr virus (EBV) has been identified as a putative trigger of multiple sclerosis (MS). Previously, we reported that mice latently infected with murine gammaherpesvirus 68 (γHV-68), the murine homolog to EBV, and induced for experimental autoimmune encephalomyelitis (EAE), developed an enhanced disease more reminiscent of MS. These prior results showed that expression of CD40 on CD11b+CD11c+ cells in latently infected mice was required to prime the strong Th1 response driving disease as well as decreasing Treg frequencies in the periphery and CNS. Subsequent work demonstrated that transfer of B cells from latently infected mice was sufficient to enhance disease. Herein, we show that B cells from infected mice do not need type I IFN signaling to drive a strong Th1 response, yet are important in driving infiltration of the CNS by CD8+ T cells. Given the importance of type I IFNs in MS, we used IFNARko mice in order to determine if type I IFN signaling was important in the enhancement of EAE in latently infected mice. We found that while type I IFNs are important for the control of γHV-68 infection and maintenance of latency, they do not have a direct effect in the development of enhanced EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Gammaherpesvirinae , Interferón Tipo I , Animales , Linfocitos T CD8-positivos , Encefalomielitis Autoinmune Experimental/inmunología , Infección Latente/inmunología , Infección Latente/virología , Ratones
5.
Endocrinology ; 163(11)2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36048448

RESUMEN

A central goal of physiological research is the understanding of cell-specific roles of disease-associated genes. Cre-mediated recombineering is the tool of choice for cell type-specific analysis of gene function in preclinical models. In the type 1 diabetes (T1D) research field, multiple lines of nonobese diabetic (NOD) mice have been engineered to express Cre recombinase in pancreatic ß cells using insulin promoter fragments, but tissue promiscuity remains a concern. Constitutive Ins1tm1.1(cre)Thor (Ins1Cre) mice on the C57/bl6-J background have high ß-cell specificity with no reported off-target effects. We explored whether NOD:Ins1Cre mice could be used to investigate ß-cell gene deletion in T1D disease modeling. We studied wild-type (Ins1WT/WT), Ins1 heterozygous (Ins1Cre/WT or Ins1Neo/WT), and Ins1 null (Ins1Cre/Neo) littermates on a NOD background. Female Ins1Neo/WT mice exhibited significant protection from diabetes, with further near-complete protection in Ins1Cre/WT mice. The effects of combined neomycin and Cre knockin in Ins1Neo/Cre mice were not additive to the Cre knockin alone. In Ins1Neo/Cre mice, protection from diabetes was associated with reduced insulitis at age 12 weeks. Collectively, these data confirm previous reports that loss of Ins1 alleles protects NOD mice from diabetes development and demonstrates, for the first time, that Cre itself may have additional protective effects. This has important implications for the experimental design and interpretation of preclinical T1D studies using ß-cell-selective Cre in NOD mice.


Asunto(s)
Diabetes Mellitus Tipo 1 , Dosificación de Gen , Células Secretoras de Insulina , Insulina , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/prevención & control , Femenino , Insulina/genética , Células Secretoras de Insulina/metabolismo , Integrasas , Ratones , Ratones Endogámicos NOD , Neomicina/metabolismo
6.
Sci Adv ; 8(47): eade6844, 2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36427301

RESUMEN

While age-associated B cells (ABCs) are known to expand and persist following viral infection and during autoimmunity, their interactions are yet to be studied together in these contexts. Here, we directly compared CD11c+T-bet+ ABCs using models of Epstein-Barr virus (EBV), gammaherpesvirus 68 (γHV68), multiple sclerosis (MS), and experimental autoimmune encephalomyelitis (EAE), and found that each drives the ABC population to opposing phenotypes. EBV infection has long been implicated in MS, and we have previously shown that latent γHV68 infection exacerbates EAE. Here, we demonstrate that ABCs are required for γHV68-enhanced disease. We then show that the circulating ABC population is expanded and phenotypically altered in people with relapsing MS. In this study, we show that viral infection and autoimmunity differentially affect the phenotype of ABCs in humans and mice, and we identify ABCs as functional mediators of viral-enhanced autoimmunity.

7.
Front Immunol ; 12: 751341, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804036

RESUMEN

Seemingly redundant in function, melanoma differentiation-associated protein 5 (MDA5) and toll-like receptor- 3 (TLR3) both sense RNA viruses and induce type I interferon (IFN-I). Herein, we demonstrate that changes in sensing of the same virus by MDA5 and TLR3 can lead to distinct signatures of IFN-α and IFN-ß resulting in different disease outcomes. Specifically, infection with a diabetogenic islet ß cell-tropic strain of coxsackievirus (CB4) results in diabetes protection under reduced MDA5 signaling conditions while reduced TLR3 function retains diabetes susceptibility. Regulating the induction of IFN-I at the site of virus infection creates a local site of interferonopathy leading to loss of T cell regulation and induction of autoimmune diabetes. We have not demonstrated another way to prevent T1D in the NOD mouse, rather we believe this work has provided compounding evidence for a specific control of IFN-I to drive a myriad of responses ranging from virus clearance to onset of autoimmune diabetes.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Citocinas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Helicasa Inducida por Interferón IFIH1/inmunología , Receptor Toll-Like 3/inmunología , Animales , Enterovirus Humano B , Femenino , Helicasa Inducida por Interferón IFIH1/genética , Masculino , Ratones Endogámicos NOD , Ratones Transgénicos , Receptor Toll-Like 3/genética
8.
Elife ; 102021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34080972

RESUMEN

Epstein-Barr virus (EBV) infection is associated with rheumatoid arthritis (RA) in adults, though the nature of the relationship remains unknown. Herein, we have examined the contribution of viral infection to the severity of arthritis in mice. We have provided the first evidence that latent gammaherpesvirus infection enhances clinical arthritis, modeling EBV's role in RA. Mice latently infected with a murine analog of EBV, gammaherpesvirus 68 (γHV68), develop more severe collagen-induced arthritis and a Th1-skewed immune profile reminiscent of human disease. We demonstrate that disease enhancement requires viral latency and is not due to active virus stimulation of the immune response. Age-associated B cells (ABCs) are associated with several human autoimmune diseases, including arthritis, though their contribution to disease is not well understood. Using ABC knockout mice, we have provided the first evidence that ABCs are mechanistically required for viral enhancement of disease, thereby establishing that ABCs are impacted by latent gammaherpesvirus infection and provoke arthritis.


Rheumatoid arthritis is one of the most common autoimmune diseases, leaving patients in pain as their immune system mistakenly attacks the lining of their joints. The precise cause is unknown, but research suggests a link to the Epstein-Barr virus, the agent responsible for mononucleosis (also known as glandular fever). After infection and recovery, the virus remains in the body, lying dormant inside immune 'B cells' which are often responsible for autoimmune diseases. Of particular interest are a sub-group known as 'age-associated B-cells', which are mostly cells left over from fighting past infections such as mononucleosis. Yet, the link between Epstein-Barr virus and rheumatoid arthritis remains hard to investigate because of the long gap between the two diseases: the virus mostly affects children and young people, while rheumatoid arthritis tends to develop in middle age. To investigate how exactly the two conditions are connected, Mouat et al. created a new animal model: they infected young mice with the murine equivalent of the Epstein-Barr virus, and then used a collagen injection to trigger rheumatoid arthritis-like disease once the animals were older. Next, Mouat et al. monitored the paws of the mice, revealing that viral infection early in life worsened arthritis later on. These animals also had more age-associated B cells than normal, and the cells showed signs of participating in inflammation. On the other hand, early viral infection did not make arthritis worse in mice unable to produce age-associated B cells. Taken together, these results suggest that the immune cells are required to enhance the effect of the viral infection on rheumatoid arthritis. This new insight may help to refine current treatments that work by reducing the overall number of B cells. Ultimately, the animal model developed by Mouat et al. could be useful to identify better ways to diagnose, monitor and treat this debilitating disease.


Asunto(s)
Artritis Experimental/virología , Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/patogenicidad , Infección Latente/virología , Latencia del Virus , Factores de Edad , Animales , Antígenos CD19/genética , Antígenos CD19/metabolismo , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Estudios de Casos y Controles , Citocinas/metabolismo , Progresión de la Enfermedad , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/metabolismo , Femenino , Herpesvirus Humano 4/inmunología , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Infección Latente/inmunología , Infección Latente/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Índice de Severidad de la Enfermedad , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/virología
9.
Front Immunol ; 11: 584297, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329556

RESUMEN

Multiple sclerosis (MS) is caused by a combination of genetic and environmental factors. It is believed that previous infection with Epstein Barr Virus (EBV) plays an important role in the development of MS. Previously, we developed a murine model where latent infection with gamma herpesvirus 68 (γHV-68), a murine homolog to EBV, enhanced the symptoms of experimental autoimmune encephalomyelitis (EAE), resulting in disease that more closely resembles MS in humans. Here, we explored the conditions that were necessary for EAE enhancement. We showed that latently infected CD19+IgD- B cells were capable of enhancing EAE symptoms when transferred from mice previously infected with γHV-68 into uninfected mice. We also observed a prevention of enhancement when B cells were depleted before infection. However, depletion after the establishment of latency only partially reduced EAE. This indicated the existence of a mechanism where B cells play an important role as antigen presenting cells (APCs) prior to EAE induction for the priming of Th1 cells. It is possible that these signals persist even after B cell depletion, strongly suggesting a paracrine signaling modulation of non-B cell APCs. These results strongly support the concept that EBV contributes to the development of autoimmunity and highlights the need for a vaccine against EBV that could limit or prevent multiple sclerosis development.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Animales , Células Presentadoras de Antígenos/inmunología , Autoinmunidad/inmunología , Encéfalo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/virología , Ratones , Ratones Endogámicos C57BL , Comunicación Paracrina/inmunología , Transducción de Señal/inmunología , Células TH1/inmunología
10.
Microorganisms ; 8(12)2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33291747

RESUMEN

The current methods to study the distribution and dynamics of viral RNA molecules inside infected cells are not ideal, as electron microscopy and immunohistochemistry can only detect mature virions, and quantitative real-time PCR does not reveal localized distribution of RNAs. We demonstrated here the branched DNA in situ hybridization (bDNA ISH) technology to study both the amount and location of the emerging -RNA and +RNA during acute and persistent enterovirus infections. According to our results, the replication of the viral RNA started 2-3 h after infection and the translation shortly after at 3-4 h post-infection. The replication hotspots with newly emerging -RNA were located quite centrally in the cell, while the +RNA production and most likely virion assembly took place in the periphery of the cell. We also discovered that the pace of replication of -RNA and +RNA strands was almost identical, and -RNA was absent during antiviral treatments. ViewRNA ISH with our custom probes also showed a good signal during acute and persistent enterovirus infections in cell and mouse models. Considering these results, along with the established bDNA FISH protocol modified by us, the effects of antiviral drugs and the emergence of enterovirus RNAs in general can be studied more effectively.

11.
J Thromb Haemost ; 17(3): 482-491, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30659719

RESUMEN

Essentials The coagulation initiator, tissue factor (TF), is on the herpes simplex virus 1 (HSV1) surface. HSV1 surface TF was examined in mice as an antiviral target since it enhances infection in vitro. HSV1 surface TF facilitated infection of all organs evaluated and anticoagulants were antiviral. Protease activated receptor 2 inhibited infection in vivo and its pre-activation was antiviral. SUMMARY: Background Tissue factor (TF) is the essential cell surface initiator of coagulation, and mediates cell signaling through protease-activated receptor (PAR) 2. Having a diverse cellular distribution, TF is involved in many biological pathways and pathologies. Our earlier work identified host cell-derived TF on the envelope covering several viruses, and showed its involvement in enhanced cell infection in vitro. Objective In the current study, we evaluated the in vivo effects of virus surface TF on infection and on the related modulator of infection PAR2. Methods With the use of herpes simplex virus type 1 (HSV1) as a model enveloped virus, purified HSV1 was generated with or without envelope TF through propagation in a TF-inducible cell line. Infection was studied after intravenous inoculation of BALB/c, C57BL/6J or C57BL/6J PAR2 knockout mice with 5 × 105 plaque-forming units of HSV1, mimicking viremia. Three days after inoculation, organs were processed, and virus was quantified with plaque-forming assays and quantitative real-time PCR. Results Infection of brain, lung, heart, spinal cord and liver by HSV1 required viral TF. Demonstrating promise as a therapeutic target, virus-specific anti-TF mAbs or small-molecule inhibitors of coagulation inhibited infection. PAR2 modulates HSV1 in vivo as demonstrated with PAR2 knockout mice and PAR2 agonist peptide. Conclusion TF is a constituent of many permissive host cell types. Therefore, the results presented here may explain why many viruses are correlated with hemostatic abnormalities, and indicate that TF is a novel pan-specific envelope antiviral target.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1/metabolismo , Tromboplastina/administración & dosificación , Proteínas del Envoltorio Viral/administración & dosificación , Animales , Anticoagulantes/farmacología , Antivirales/farmacología , Modelos Animales de Enfermedad , Femenino , Herpes Simple/sangre , Herpes Simple/tratamiento farmacológico , Herpes Simple/inmunología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno , Inyecciones Intravenosas , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Células TH1/inmunología , Células TH1/virología , Tromboplastina/metabolismo , Proteínas del Envoltorio Viral/metabolismo
12.
Front Immunol ; 10: 2473, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31736943

RESUMEN

Calcium (Ca2+) is a vital secondary messenger in T lymphocytes regulating a vast array of important events including maturation, homeostasis, activation, and apoptosis and can enter the cell through CRAC, TRP, and CaV channels. Here we describe a mutation in the L-type Ca2+ channel CaV1.4 leading to T lymphocyte dysfunction, including several hallmarks of immunological exhaustion. CaV1.4-deficient mice exhibited an expansion of central and effector memory T lymphocytes, and an upregulation of inhibitory receptors on several T cell subsets. Moreover, the sustained elevated levels of activation markers on B lymphocytes suggest that they are in a chronic state of activation. Functionally, T lymphocytes exhibited a reduced store-operated Ca2+ flux compared to wild-type controls. Finally, modifying environmental conditions by herpes virus infection exacerbated the dysfunctional immune phenotype of the CaV1.4-deficient mice. This is the first example where the mutation of a CaV channel leads to T lymphocyte dysfunction, including the upregulation of several inhibitory receptors, hallmarks of T cell exhaustion, and establishes the physiological importance of CaV channel signaling in maintaining a nimble immune system.


Asunto(s)
Canales de Calcio Tipo L/genética , Mutación , Fenotipo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Relación CD4-CD8 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Calcio/metabolismo , Señalización del Calcio , Expresión Génica , Estudios de Asociación Genética , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/virología , Memoria Inmunológica , Inmunofenotipificación , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Ratones , Ratones Noqueados , Virus de la Hepatitis Murina/inmunología
13.
Sci Rep ; 8(1): 4588, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29545618

RESUMEN

Pancreatic beta-cells are selectively destroyed by the host immune system in type 1 diabetes. Thus, drugs that preserve beta-cell mass and/or function have the potential to prevent or slow the progression of this disease. We recently reported that the use-dependent sodium channel blocker, carbamazepine, protects beta-cells from inflammatory cytokines in vitro. Here, we tested the effects of carbamazepine treatment in female non-obese diabetic (NOD) mice by supplementing LabDiet 5053 with 0.5% w/w carbamazepine to achieve serum carbamazepine levels of 14.98 ± 3.19 µM. Remarkably, diabetes incidence over 25 weeks, as determined by fasting blood glucose, was ~50% lower in carbamazepine treated animals. Partial protection from diabetes in carbamazepine-fed NOD mice was also associated with improved glucose tolerance at 6 weeks of age, prior to the onset of diabetes in our colony. Less insulitis was detected in carbamazepine treated NOD mice at 6 weeks of age, but we did not observe differences in CD4+ and CD8+ T cell composition in the pancreatic lymph node, as well as circulating markers of inflammation. Taken together, our results demonstrate that carbamazepine reduces the development of type 1 diabetes in NOD mice by maintaining functional beta-cell mass.


Asunto(s)
Carbamazepina/uso terapéutico , Diabetes Mellitus Tipo 1/prevención & control , Bloqueadores de los Canales de Sodio/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Carbamazepina/sangre , Carbamazepina/farmacología , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/veterinaria , Femenino , Prueba de Tolerancia a la Glucosa , Incidencia , Células Secretoras de Insulina/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos NOD , Bloqueadores de los Canales de Sodio/sangre , Bloqueadores de los Canales de Sodio/farmacología
14.
Diabetes ; 64(6): 2184-93, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25591872

RESUMEN

Although it is widely accepted that type 1 diabetes (T1D) is the result of the autoimmune destruction of insulin-producing ß-cells in the pancreas, little is known about the events leading to islet autoimmunity. Epidemiological and genetic data have associated virus infections and antiviral type I interferon (IFN-I) response genes with T1D. Genetic variants in the T1D risk locus interferon induced with helicase C domain 1 (IFIH1) have been identified by genome-wide association studies to confer resistance to T1D and result in the reduction in expression of the intracellular RNA virus sensor known as melanoma differentiation-associated protein 5 (MDA5). Here, we translate the reduction in IFIH1 gene expression that results in protection from T1D. Our functional studies demonstrate that mice heterozygous at the Ifih1 gene express less than half the level of MDA5 protein, which leads to a unique antiviral IFN-I signature and adaptive response after virus infection that protects from T1D. IFIH1 heterozygous mice have a regulatory rather than effector T-cell response at the site of autoimmunity, supporting IFIH1 expression as an essential regulator of the diabetogenic T-cell response and providing a potential mechanism for patients carrying IFIH1 protective polymorphisms.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Animales , ARN Helicasas DEAD-box/genética , Diabetes Mellitus Tipo 1/genética , Femenino , Citometría de Flujo , Predisposición Genética a la Enfermedad/genética , Heterocigoto , Inmunohistoquímica , Helicasa Inducida por Interferón IFIH1 , Masculino , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Sci Rep ; 5: 13995, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26356194

RESUMEN

Epstein-Barr virus (EBV) has been identified as a putative environmental trigger of multiple sclerosis (MS) by multiple groups working worldwide. Previously, we reported that when experimental autoimmune encephalomyelitis (EAE) was induced in mice latently infected with murine γ-herpesvirus 68 (γHV-68), the murine homolog to EBV, a disease more reminiscent of MS developed. Specifically, MS-like lesions developed in the brain that included equal numbers of IFN-γ producing CD4(+) and CD8(+) T cells and demyelination, none of which is observed in MOG induced EAE. Herein, we demonstrate that this enhanced disease was dependent on the γHV-68 latent life cycle and was associated with STAT1 and CD40 upregulation on uninfected dendritic cells. Importantly, we also show that, during viral latency, the frequency of regulatory T cells is reduced via a CD40 dependent mechanism and this contributes towards a strong T helper 1 response that resolves in severe EAE disease pathology. Latent γ-herpesvirus infection established a long-lasting impact that enhances subsequent adaptive autoimmune responses.


Asunto(s)
Autoinmunidad/genética , Antígenos CD40/genética , Expresión Génica , Esclerosis Múltiple/etiología , Rhadinovirus/fisiología , Latencia del Virus , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental , Infecciones por Herpesviridae/complicaciones , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/fisiología , Humanos , Inmunofenotipificación , Ratones , Esclerosis Múltiple/patología , Fenotipo , Factor de Transcripción STAT1/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Regulación hacia Arriba
16.
PLoS One ; 10(12): e0145179, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26674203

RESUMEN

Autoimmune destruction of the pancreatic islets in Type 1 diabetes is mediated by both increased proinflammatory (Teff) and decreased regulatory (Treg) T lymphocytes resulting in a significant decrease in the Treg:Teff ratio. The non-obese diabetic (NOD) mouse is an excellent in vivo model for testing potential therapeutics for attenuating the decrease in the Treg:Teff ratio and inhibiting disease pathogenesis. Here we show for the first time that a bioreactor manufactured therapeutic consisting of a complex of miRNA species (denoted as TA1) can effectively reset the NOD immune system from a proinflammatory to a tolerogenic state thus preventing or delaying autoimmune diabetes. Treatment of NOD mice with TA1 resulted in a systemic broad-spectrum upregulation of tolerogenic T cell subsets with a parallel downregulation of Teff subsets yielding a dramatic increase in the Treg:Teff ratio. Moreover, the murine-derived TA1 was highly effective in the inhibition of allorecognition of HLA-disparate human PBMC. TA1 demonstrated dose-responsiveness and exhibited equivalent or better inhibition of allorecognition driven proliferation than etanercept (a soluble TNF receptor). These findings demonstrate that miRNA-based therapeutics can effectively attenuate or arrest autoimmune disease processes and may be of significant utility in a broad range of autoimmune diseases including Type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Tratamiento con ARN de Interferencia , Animales , Ratones , Ratones Endogámicos NOD , Linfocitos T Reguladores/inmunología
17.
Autoimmunity ; 46(1): 62-73, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23039284

RESUMEN

During the progression of autoimmune (type 1) diabetes, T cells and macrophages infiltrate the pancreas, disrupt islet function, and destroy insulin-producing beta cells. B-lymphocytes, particularly innate like B-cell populations such as marginal zone B cells and B-1 cells, have been implicated in many autoimmune diseases, and non-obese diabetic (NOD) mice that lack B cells do not develop spontaneous autoimmune diabetes. Hence, inhibitors of B cell signaling pathways could be useful for limiting the autoimmune processes that contribute to type 1 diabetes. Signaling via phosphoinositide 3-kinase (PI3K) regulates many cellular processes. The p110δ isoform of PI3K is expressed primarily in cells of hematopoietic origin and the catalytic activity of p110δ is important for B cell migration, activation, proliferation, and antigen presentation. Because innate-like B cells are particularly sensitive to inhibition of p110δ activity, and p110δ inhibitors also suppress pro-inflammatory functions of other cell types that contribute to autoimmunity, we tested whether a p110δ inhibitor could delay the onset or reduce the incidence of autoimmune diabetes in NOD mice. We found that long-term preventative treatment of pre-diabetic NOD mice with IC87114, a highly selective small molecule inhibitor of p110δ, reduced the infiltration of inflammatory cells into the pancreatic islets and, accordingly, delayed and reduced the loss of glucose homeostasis. Moreover in a therapeutic treatment mode, IC87114 treatment conferred prolonged protection from progression to overt diabetes in a number of animals. These findings suggest that PI3Kδ inhibitors could be useful for managing type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/enzimología , Células Secretoras de Insulina/inmunología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autoinmunidad/inmunología , Linfocitos B/enzimología , Linfocitos B/inmunología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Histocitoquímica , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Estadísticas no Paramétricas
18.
PLoS One ; 8(6): e65965, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840383

RESUMEN

The HIV/AIDS pandemic remains an enormous global health concern. Despite effective prevention options, 2.6 million new infections occur annually, with women in developing countries accounting for more than half of these infections. New prevention strategies that can be used by women are urgently needed. Topical microbicides specific for HIV-1 represent a promising prevention strategy. Conceptually, using harmless bacteria to display peptides or proteins capable of blocking entry provides an inexpensive approach to microbicide development. To avoid the potential pitfalls of engineering commensal bacteria, our strategy is to genetically display infection inhibitors on a non-native bacterium and rely on topical application of stabilized bacteria before potential virus exposure. Due to the high density cell-surface display capabilities and the inherent low toxicity of the bacterium, the S-layer mediated protein display capabilities of the non-pathogenic bacterium Caulobacter crescentus has been exploited for this approach. We have demonstrated that C. crescentus displaying MIP1α or CD4 interfered with the virus entry pathway and provided significant protection from HIV-1 pseudovirus representing clade B in a standard single cycle infection assay. Here we have expanded our C. crescentus based microbicide approach with additional and diverse classes of natural and synthetic inhibitors of the HIV-1 entry pathway. All display constructs provided variable but significant protection from HIV-1 infection; some with protection as high as 70%. Further, we describe protection from infection with additional viral clades. These findings indicate the significant potential for engineering C. crescentus to be an effective and readily adaptable HIV-1 microbicide platform.


Asunto(s)
Antígenos CD4/farmacología , Caulobacter crescentus/genética , Quimiocina CCL3/farmacología , VIH-1/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Administración Tópica , Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/uso terapéutico , Antiinfecciosos Locales/farmacología , Antiinfecciosos Locales/uso terapéutico , Antígenos CD4/genética , Antígenos CD4/uso terapéutico , Caulobacter crescentus/metabolismo , Quimiocina CCL3/genética , Quimiocina CCL3/uso terapéutico , Femenino , Ingeniería Genética , Infecciones por VIH/prevención & control , Infecciones por VIH/terapia , VIH-1/genética , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo
19.
PLoS One ; 7(2): e31153, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355341

RESUMEN

Progression towards type 1 diabetes (T1D) in susceptible patients is linked to a progressive decline in the capacity of regulatory T cells (Treg) to maintain tolerance. As such, therapies aimed at redressing the failing Treg compartment have been the subject of intense investigation. Treg dysfunction in T1D has recently been linked to a reduced capacity of antigen presenting cells (APCs) to maintain Treg function rather than Treg intrinsic defects. This suggests that therapies aimed simply at addressing the failing Treg compartment are unlikely to provide long-term protection. Here, we demonstrate that modulation of the inflammatory status of CD11b+CD11c- APCs favors the upregulation of protective Tregs in a mouse model of T1D. We further demonstrate that reduced expression of the costimulatory molecule CD40 plays a role in this increased immunoregulatory capacity. Strikingly, Treg upregulation resulted exclusively from an increase in natural Tregs rather than the peripheral conversion of conventional T cells. This suggests that modulation of CD11b+ CD11c- APCs inflammatory properties favors the establishment of natural Treg responses that, unlike adaptive Treg responses, are likely to maintain tolerance to a broad range of antigens. As such, modulation of this APC subset represents a potential therapeutic avenue to reestablish peripheral tolerance and protect from autoimmune diseases such as T1D.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Tolerancia Inmunológica/inmunología , Inmunomodulación/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígeno CD11b/metabolismo , Diabetes Mellitus Tipo 1/virología , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Inflamación/inmunología , Inflamación/prevención & control , Inflamación/virología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
20.
PLoS One ; 5(4): e10366, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20442778

RESUMEN

The development of alternative strategies to prevent HIV infection is a global public health priority. Initial efforts in anti-HIV microbicide development have met with poor success as the strategies have relied on a non-specific mechanism of action. Here, we report the development of a microbicide aimed at specifically blocking HIV entry by displaying molecular components of the HIV/host cell attachment complex on the surface of Caulobacter crescentus, a harmless aquatic bacterium. This bacterium can be readily manipulated to present heterologous proteins at high density on its surface by genetic insertion into its crystalline surface layer protein. In separate constructions, we generated bacteria displaying domain 1 of CD4 and MIP1alpha. Each moiety reacted with specific antibodies by Western immunoblot and immuno-fluorescence microscopy. Microbicide functionality was assessed using an HIV pseudotype virus assay system representing Clade B subtypes. Bacteria displaying MIP1alpha reduced infectivity by 35-78% depending on the specific subtype while CD4 display reduced infection by as much as 56%. Combinations of both constructs reduced infectivity by nearly 98%. We demonstrated that HIV infection could be inhibited using a strategy aimed at HIV-specific molecular interactions with Caulobacter surface protein display, and that sufficient protein folding and conformation could be mimicked to bind and block entry. Further, this is the first demonstration that Caulobacter surface protein display may be a useful approach to preventing HIV infection or other viruses as a microbicide. We propose that this harmless bacterium, which is inexpensive to produce and formulate, might be suitable for topical applications as a viable alternative in the search for effective microbicides to counteract the world wide incidence of HIV infection.


Asunto(s)
Fármacos Anti-VIH , Antígenos CD4/uso terapéutico , Caulobacter crescentus/genética , Quimiocina CCL3/uso terapéutico , Antiinfecciosos , Antígenos CD4/genética , Quimiocina CCL3/genética , Técnicas de Transferencia de Gen , Infecciones por VIH/prevención & control , Infecciones por VIH/terapia , VIH-1 , Humanos , Inmunoensayo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA