Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunogenetics ; 75(1): 27-37, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36097289

RESUMEN

We previously reported that nonobese diabetic (NOD) congenic mice (NOD.c3c4 mice) developed an autoimmune biliary disease (ABD) with similarities to human primary biliary cholangitis (PBC), including anti-mitochondrial antibodies and organ-specific biliary lymphocytic infiltrates. We narrowed the possible contributory regions in a novel NOD.Abd3 congenic mouse to a B10 congenic region on chromosome 1 ("Abd3") and a mutated Pkhd1 gene (Pkhd1del36-67) upstream from Abd3, and we showed via backcrossing studies that the NOD genetic background was necessary for disease. Here, we show that NOD.Abd3 mice develop anti-PDC-E2 autoantibodies at high levels, and that placing the chromosome 1 interval onto a scid background eliminates disease, demonstrating the critical role of the adaptive immune system in pathogenesis. While the NOD genetic background is essential for disease, it was still unclear which of the two regions in the Abd3 locus were necessary and sufficient for disease. Here, using a classic recombinant breeding approach, we prove that the mutated Pkhd1del36-67 alone, on the NOD background, causes ABD. Further characterization of the mutant sequence demonstrated that the Pkhd1 gene is disrupted by an ETnII-beta retrotransposon inserted in intron 35 in an anti-sense orientation. Homozygous Pkhd1 mutations significantly affect viability, with the offspring skewed away from a Mendelian distribution towards NOD Pkhd1 homozygous or heterozygous genotypes. Cell-specific abnormalities, on a susceptible genetic background, can therefore induce an organ-specific autoimmunity directed to the affected cells. Future work will aim to characterize how mutant Pkhd1 can cause such an autoimmune response.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus , Ratones , Animales , Humanos , Ratones Endogámicos NOD , Autoanticuerpos/genética , Ratones Congénicos , Antecedentes Genéticos , Diabetes Mellitus Tipo 1/genética , Ratones Endogámicos C57BL , Receptores de Superficie Celular/genética
2.
J Autoimmun ; 139: 103070, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37390745

RESUMEN

Infectious diseases are commonly implicated as potential initiators of autoimmune diseases (ADs) and represent the most commonly known factor in the development of autoimmunity in susceptible individuals. Epidemiological data and animal studies on multiple ADs suggest that molecular mimicry is one of the likely mechanisms for the loss of peripheral tolerance and the development of clinical disease. Besides molecular mimicry, other mechanisms such as defects in central tolerance, nonspecific bystander activation, epitope-determinant spreading, and/or constant antigenic stimuli, may also contribute for breach of tolerance and to the development of ADs. Linear peptide homology is not the only mechanism by which molecular mimicry is established. Peptide modeling (i.e., 3D structure), molecular docking analyses, and affinity estimation for HLAs are emerging as critical strategies when studying the links of molecular mimicry in the development of autoimmunity. In the current pandemic, several reports have confirmed an influence of SARS-CoV-2 on subsequent autoimmunity. Bioinformatic and experimental evidence support the potential role of molecular mimicry. Peptide dimensional analysis requires more research and will be increasingly important for designing and distributing vaccines and better understanding the role of environmental factors related to autoimmunity.


Asunto(s)
Enfermedades Autoinmunes , COVID-19 , Animales , Autoinmunidad , Imitación Molecular , Simulación del Acoplamiento Molecular , SARS-CoV-2 , Enfermedades Autoinmunes/epidemiología
3.
J Autoimmun ; 141: 103114, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37748979

RESUMEN

The gastrointestinal tract is home to the largest microbial population in the human body. The gut microbiota plays significant roles in the development of the gut immune system and has a substantial impact on the maintenance of immune tolerance beginning in early life. These microbes interact with the immune system in a dynamic and interdependent manner. They generate immune signals by presenting a vast repertoire of antigenic determinants and microbial metabolites that influence the development, maturation and maintenance of immunological function and homeostasis. At the same time, both the innate and adaptive immune systems are involved in modulating a stable microbial ecosystem between the commensal and pathogenic microorganisms. Hence, the gut microbial population and the host immune system work together to maintain immune homeostasis synergistically. In susceptible hosts, disruption of such a harmonious state can greatly affect human health and lead to various auto-inflammatory and autoimmune disorders. In this review, we discuss our current understanding of the interactions between the gut microbiota and immunity with an emphasis on: a) important players of gut innate and adaptive immunity; b) the contribution of gut microbial metabolites; and c) the effect of disruption of innate and adaptive immunity as well as alteration of gut microbiome on the molecular mechanisms driving autoimmunity in various autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Microbioma Gastrointestinal , Humanos , Ecosistema , Sistema Inmunológico , Inmunidad Adaptativa , Tolerancia Inmunológica , Disbiosis
4.
Hepatology ; 75(2): 266-279, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34608663

RESUMEN

BACKGROUND AND AIMS: The increased frequency of urinary tract infections in patients with primary biliary cholangitis (PBC) and the cross-reactivity between the lipoyl domains (LD) of human pyruvate dehydrogenase complex (hPDC-E2) and Escherichia coli PDC-E2 (ePDC-E2) have long suggested a role of E. coli in causality of PBC. This issue, however, has remained speculative. We hypothesized that by generating specific constructs of human and E. coli PDC-E2, we would be able to assess the specificity of autoantibody responses and define whether exposure to E. coli in susceptible hosts is the basis for the antimitochondrial antibody (AMA) response. APPROACH AND RESULTS: Importantly, the reactivity of hPDC-E2 LD (hPDC-E2LD) affinity-purified antibodies against hPDC-E2LD could only be removed by prior absorption with hPDC-E2LD and not ePDC-E2, suggesting the presence of unique human PDC-E2 epitopes distinct from E. coli PDC-E2. To identify the autoepitope(s) present in hPDC-E2LD, a more detailed study using a variety of PDC-E2 constructs was tested, including the effect of lipoic acid (LA) on ePDC-E2 conformation and AMA recognition. Individual recombinant ePDCE2 LD domains LD1, LD2 and LD3 did not react with either AMA or antibodies to LA (anti-LA), but in contrast, anti-LA was readily reactive against purified recombinant LD1, LD2, and LD3 expressed in tandem (LP); such reactivity increased when LP was precultured with LA. Moreover, when the three LD (LD1, LD2, LD3) domains were expressed in tandem in pET28a or when LD1 was expressed in another plasmid pGEX, they were lipoylated and reactive to PBC sera. CONCLUSIONS: In conclusion, our data are consistent with an exposure to E. coli that elicits specific antibody to ePDC-E2 resulting in determinant spreading and the classic autoantibody to hPDC-E2LD. We argue this is the first step to development of human PBC.


Asunto(s)
Autoantígenos/inmunología , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/inmunología , Infecciones por Escherichia coli/complicaciones , Escherichia coli/inmunología , Cirrosis Hepática Biliar/microbiología , Mitocondrias/inmunología , Proteínas Mitocondriales/inmunología , Autoanticuerpos/sangre , Estudios de Casos y Controles , Reacciones Cruzadas/inmunología , Epítopos/inmunología , Escherichia coli/enzimología , Hepatitis Autoinmune/sangre , Humanos , Lipoilación , Conformación Molecular/efectos de los fármacos , Ácido Tióctico/inmunología , Ácido Tióctico/farmacología
5.
J Immunol ; 207(8): 2051-2059, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526376

RESUMEN

Ag-specific immunotherapy to restore immune tolerance to self-antigens, without global immune suppression, is a long-standing goal in the treatment of autoimmune disorders such as type 1 diabetes (T1D). However, vaccination with autoantigens such as insulin or glutamic acid decarboxylase have largely failed in human T1D trials. Induction and maintenance of peripheral tolerance by vaccination requires efficient autoantigen presentation by APCs. In this study, we show that a lipophilic modification at the N-terminal end of CD4+ epitopes (lipo-peptides) dramatically improves peptide Ag presentation. We designed amphiphilic lipo-peptides to efficiently target APCs in the lymph nodes by binding and trafficking with endogenous albumin. Additionally, we show that lipophilic modification anchors the peptide into the membranes of APCs, enabling a bivalent cell-surface Ag presentation. The s.c. injected lipo-peptide accumulates in the APCs in the lymph node, enhances the potency and duration of peptide Ag presentation by APCs, and induces Ag-specific immune tolerance that controls both T cell- and B cell-mediated immunity. Immunization with an amphiphilic insulin B chain 9-23 peptide, an immunodominant CD4+ T cell epitope in NOD mice, significantly suppresses the activation of T cells, increases inhibitory cytokine production, induces regulatory T cells, and delays the onset and lowers the incidence of T1D. Importantly, treatment with a lipophilic ß-cell peptide mixture delays progression to end-stage diabetes in acutely diabetic NOD mice, whereas the same doses of standard soluble peptides were not effective. Amphiphilic modification effectively enhances Ag presentation for peptide-based immune regulation of autoimmune diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/metabolismo , Insulina/metabolismo , Fragmentos de Péptidos/metabolismo , Tensoactivos/metabolismo , Albúminas , Animales , Presentación de Antígeno , Femenino , Humanos , Tolerancia Inmunológica , Inmunización , Inmunomodulación , Insulina/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Fragmentos de Péptidos/inmunología
6.
J Autoimmun ; 132: 102897, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36029718

RESUMEN

OBJECTIVE: The ability to regulate B cell development has long been recognized to have therapeutic potential in a variety of autoimmune diseases. However, despite the presence of a classic autoantibody in primary biliary cholangitis (PBC), B cell depleting therapy and indeed therapy with other biologic agents has been disappointing. Unsuccessful treatment using Rituximab is associated with elevation of B-cell activating factor (BAFF) level. Indeed, therapies for PBC remain directed at modulating bile salt biology, rather than targeting effector pathways. With these data in mind, we proposed that targeting two major stages of B cell development, namely long-lived memory B cells and short-lived peripheral autoreactive plasma cells would have therapeutic potential. METHODS: To address this thesis, we administrated anti-BAFF and anti-CD20 monoclonal antibody to ARE-Del mice, a well-characterized murine model of human PBC. We evaluated and compared the therapeutic efficacy of the two agents individually and the combination of anti-BAFF and anti-CD20 in female mice with well-established disease. RESULTS: Our data demonstrate that there was an increased level of B cell depletion that resulted in a significantly more effective clinical and serologic response using the combination of agents as compared with the use of the individual agents. The combination of anti-BAFF and anti-CD20 treatment was more effective in reducing serum levels of antimitochondrial antibody (AMA), total IgM and IgG compared to mice treated with the 2 individual agents. Combination treatment efficiently depleted B cells in the peripheral blood, peritoneal cavity and spleen. Importantly, we identified a unique IgM+ FCRL5+ B cell subset which was sensitive to dual B-cell targeting therapy and depletion of this unique population was associated with reduced portal infiltration and bile duct damage. Taken together, our data indicate that dual B cell targeting therapy with anti-BAFF and anti-CD20 not only led to the efficient depletion of B cells both in the peripheral blood and tissues, but also led to significant clinical improvement. These findings highlight the potential application of combination of anti-BAFF and anti-CD20 in treating patients with PBC. However, additional studies in other animal models of PBC should be undertaken before considering human trials in those PBC patients who have incomplete responses to conventional therapy.


Asunto(s)
Enfermedades Autoinmunes , Colangitis , Humanos , Femenino , Ratones , Animales , Colangitis/tratamiento farmacológico , Enfermedades Autoinmunes/tratamiento farmacológico , Antígenos CD20 , Autoanticuerpos , Inmunoglobulina M
7.
Hepatology ; 74(2): 835-846, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33462854

RESUMEN

BACKGROUND AND AIMS: Primary biliary cholangitis (PBC) is a prototypical organ-specific autoimmune disease that is mediated by autoreactive T-cell attack and destruction of cholangiocytes. Despite the clear role of autoimmunity in PBC, immune-directed therapies have failed to halt PBC, including biologic therapies effective in other autoimmune diseases. MicroRNA (miRNA) dysregulation is implicated in the pathogenesis (PBC). In the dominant-negative TGF-ß receptor type II (dnTGFßRII) mouse model of PBC, autoreactive CD8 T cells play a major pathogenic role and demonstrate a striking pattern of miRNA down-regulation. Enoxacin is a small molecule fluoroquinolone that enhances miRNA biogenesis, partly by stabilizing the interaction of transactivation response RNA-binding protein with Argonaute (Ago) 2. APPROACH AND RESULTS: We hypothesized that correcting aberrant T-cell miRNA expression with enoxacin in dnTGFßRII mice could modulate autoreactive T-cell function and prevent PBC. Here, we show that liver-infiltrating dnTGFßRII CD8 T cells have significantly decreased levels of the miRNA biogenesis molecules prolyl 4-hydroxylase subunit alpha 1 (P4HA1) and Ago2 along with significantly increased levels of granzyme B and perforin. Enoxacin treatment significantly up-regulated miRNAs in dnTGFßRII CD8 T cells and effectively treated autoimmune cholangitis in dnTGFßRII mice. Enoxacin treatment directly altered T cells both ex vivo and in vitro, resulting in altered memory subset numbers, decreased proliferation, and decreased interferon-γ production. Enoxacin significantly decreased CD8 T-cell expression of the transcription factor, Runx3, and significantly decreased perforin expression at both the mRNA and protein levels. CONCLUSIONS: Enoxacin increases miRNA expression in dnTGFßRII CD8 T cells, reduces CD8 T-cell pathogenicity, and effectively halted progression of autoimmune biliary disease. Targeting the miRNA pathway is a therapeutic approach to autoimmunity that corrects pathological miRNA abnormalities in autoreactive T cells.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Enoxacino/farmacología , Cirrosis Hepática Biliar/tratamiento farmacológico , MicroARNs/biosíntesis , Linfocitos T Citotóxicos/efectos de los fármacos , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Enoxacino/uso terapéutico , Humanos , Cirrosis Hepática Biliar/genética , Cirrosis Hepática Biliar/inmunología , Ratones , Cultivo Primario de Células , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
8.
J Immunol ; 204(11): 2887-2899, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32295876

RESUMEN

CD137 modulates type 1 diabetes (T1D) progression in NOD mice. We previously showed that CD137 expression in CD4 T cells inhibits T1D, but its expression in CD8 T cells promotes disease development by intrinsically enhancing the accumulation of ß-cell-autoreactive CD8 T cells. CD137 is expressed on a subset of FOXP3+ regulatory CD4 T cells (Tregs), and CD137+ Tregs are the main source of soluble CD137. Soluble CD137 suppresses T cells in vitro by binding to the CD137 ligand (CD137L) upregulated on activated T cells. To further study how the opposing functions of CD137 are regulated, we successfully targeted Tnfsf9 (encoding CD137L) in NOD mice using the CRISPR/Cas9 system (designated NOD.Tnfsf9 -/-). Relative to wild-type NOD mice, T1D development in the NOD.Tnfsf9 -/- strain was significantly delayed, and mice developed less insulitis and had reduced frequencies of ß-cell-autoreactive CD8 T cells. Bone marrow chimera experiments showed that CD137L-deficient hematopoietic cells were able to confer T1D resistance. Adoptive T cell transfer experiments showed that CD137L deficiency on myeloid APCs was associated with T1D suppression. Conversely, lack of CD137L on T cells enhanced their diabetogenic activity. Furthermore, neither CD137 nor CD137L was required for the development and homeostasis of FOXP3+ Tregs. However, CD137 was critical for the in vivo T1D-suppressive activity of FOXP3+ Tregs, suggesting that the interaction between CD137 and CD137L regulates their function. Collectively, our results provide new insights into the complex roles of CD137-CD137L interaction in T1D.


Asunto(s)
Ligando 4-1BB/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Linfocitos T Reguladores/metabolismo , Ligando 4-1BB/genética , Animales , Antígenos CD4/metabolismo , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Factores de Transcripción Forkhead/metabolismo , Homeostasis , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Transducción de Señal , Linfocitos T Reguladores/inmunología , Quimera por Trasplante , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
9.
J Autoimmun ; 109: 102442, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32253068

RESUMEN

The Coronavirus-associated disease, that was first identified in 2019 in China (CoViD-19), is a pandemic caused by a bat-derived beta-coronavirus, named SARS-CoV2. It shares homology with SARS and MERS-CoV, responsible for past outbreaks in China and in Middle East. SARS-CoV2 spread from China where the first infections were described in December 2019 and is responsible for the respiratory symptoms that can lead to acute respiratory distress syndrome. A cytokine storm has been shown in patients who develop fatal complications, as observed in past coronavirus infections. The management includes ventilatory support and broad-spectrum antiviral drugs, empirically utilized, as a targeted therapy and vaccines have not been developed. Based upon our limited knowledge on the pathogenesis of CoViD-19, a potential role of some anti-rheumatic drugs may be hypothesized, acting as direct antivirals or targeting host immune response. Antimalarial drugs, commonly used in rheumatology, may alter the lysosomal proteases that mediates the viral entry into the cell and have demonstrated efficacy in improving the infection. Anti-IL-1 and anti-IL-6 may interfere with the cytokine storm in severe cases and use of tocilizumab has shown good outcomes in a small cohort. Baricitinib has both antiviral and anti-inflammatory properties. Checkpoints inhibitors such as anti-CD200 and anti-PD1 could have a role in the treatment of CoViD-19. Rheumatic disease patients taking immunosuppressive drugs should be recommended to maintain the chronic therapy, prevent infection by avoiding social contacts and pausing immunosuppressants in case of infection. National and international registries are being created to collect data on rheumatic patients with CoViD-19.


Asunto(s)
Terapia Biológica , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/complicaciones , Neumonía Viral/tratamiento farmacológico , Enfermedades Reumáticas/complicaciones , Enfermedades Reumáticas/terapia , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antimaláricos/uso terapéutico , Antirreumáticos/uso terapéutico , Azetidinas/uso terapéutico , Betacoronavirus/efectos de los fármacos , COVID-19 , Infecciones por Coronavirus/prevención & control , Citocinas/inmunología , Humanos , Inmunosupresores/uso terapéutico , Interleucina-1/antagonistas & inhibidores , Interleucina-6/antagonistas & inhibidores , Pandemias/prevención & control , Neumonía Viral/prevención & control , Purinas , Pirazoles , SARS-CoV-2 , Sulfonamidas/uso terapéutico , Internalización del Virus/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
10.
J Autoimmun ; 113: 102503, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32546343

RESUMEN

Glycosylation of antibodies, particularly in the Fc domain, critically modulate the ability of antibodies to bind to FcRs, maintaining immune quiescence to achieve a finely orchestrated immune response. The removal of sialic acid and galactose residues dramatically alters the physiological function of IgGs, and alterations of Ig glycosylation have been associated with several autoimmune disorders. However, Ig glycosylation has not been extensively studied in autoimmune cholangitis. We applied triple quadruple mass spectroscopy with subsequent multiple reaction monitoring to elucidate the profile, composition and linkage of sugar residues of antibody glycans in patients with primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC) and healthy controls (HC). Agalactosylated, HexNAc terminated IgG1 glycoforms were enriched in both PBC and PSC. Levels of IgM glycans at site N439 and fucosylated glycans in J chain, were significantly decreased in PBC compared to PSC and HC. PSC patients had decreased bisecting glycoforms and increased biantennary glycoforms on IgA compared to PBC. Importantly, our data demonstrate the association of distinct branching and composition patterns of Ig glycoforms with disease severity and liver cirrhosis, which highlight the importance of glycan biology as a potential mechanism and/or a disease specific signal of inflammation.


Asunto(s)
Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/diagnóstico , Colangitis Esclerosante/diagnóstico , Inmunoglobulina G/metabolismo , Cirrosis Hepática Biliar/diagnóstico , Adulto , Anciano , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/inmunología , Biomarcadores/sangre , Biomarcadores/metabolismo , Estudios de Casos y Controles , Colangitis Esclerosante/sangre , Colangitis Esclerosante/inmunología , Femenino , Glicómica/métodos , Glicosilación , Voluntarios Sanos , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Cirrosis Hepática Biliar/sangre , Cirrosis Hepática Biliar/inmunología , Masculino , Persona de Mediana Edad , Polisacáridos/inmunología , Polisacáridos/metabolismo , Índice de Severidad de la Enfermedad
11.
J Autoimmun ; 114: 102506, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32563547

RESUMEN

Coronavirus disease 2019 (COVID-19) has been categorized as evolving in overlapping phases. First, there is a viral phase that may well be asymptomatic or mild in the majority, perhaps 80% of patients. The pathophysiological mechanisms resulting in minimal disease in this initial phase are not well known. In the remaining 20% of cases, the disease may become severe and/or critical. In most patients of this latter group, there is a phase characterized by the hyperresponsiveness of the immune system. A third phase corresponds to a state of hypercoagulability. Finally, in the fourth stage organ injury and failure occur. Appearance of autoinflammatory/autoimmune phenomena in patients with COVID-19 calls attention for the development of new strategies for the management of life-threatening conditions in critically ill patients. Antiphospholipid syndrome, autoimmune cytopenia, Guillain-Barré syndrome and Kawasaki disease have each been reported in patients with COVID-19. Here we present a scoping review of the relevant immunological findings in COVID-19 as well as the current reports about autoinflammatory/autoimmune conditions associated with the disease. These observations have crucial therapeutic implications since immunomodulatory drugs are at present the most likely best candidates for COVID-19 therapy. Clinicians should be aware of these conditions in patients with COVID-19, and these observations should be considered in the current development of vaccines.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Neumonía Viral/inmunología , Inmunidad Adaptativa/genética , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/virología , Betacoronavirus/aislamiento & purificación , COVID-19 , Prueba de COVID-19 , Técnicas de Laboratorio Clínico , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/terapia , Enfermedad Crítica , Síndrome de Liberación de Citoquinas/diagnóstico , Síndrome de Liberación de Citoquinas/terapia , Síndrome de Liberación de Citoquinas/virología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Inmunidad Innata/genética , Inmunización Pasiva/métodos , Mediadores de Inflamación/sangre , Mediadores de Inflamación/inmunología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Masculino , Pandemias , Neumonía Viral/diagnóstico , Neumonía Viral/epidemiología , Neumonía Viral/terapia , Factores de Riesgo , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Factores Sexuales , Sueroterapia para COVID-19
12.
J Immunol ; 200(1): 147-162, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29158418

RESUMEN

We previously reported that NOD.c3c4 mice develop spontaneous autoimmune biliary disease (ABD) with anti-mitochondrial Abs, histopathological lesions, and autoimmune T lymphocytes similar to human primary biliary cholangitis. In this article, we demonstrate that ABD in NOD.c3c4 and related NOD ABD strains is caused by a chromosome 1 region that includes a novel mutation in polycystic kidney and hepatic disease 1 (Pkhd1). We show that a long terminal repeat element inserted into intron 35 exposes an alternative polyadenylation site, resulting in a truncated Pkhd1 transcript. A novel NOD congenic mouse expressing aberrant Pkhd1, but lacking the c3 and c4 chromosomal regions (NOD.Abd3), reproduces the immunopathological features of NOD ABD. RNA sequencing of NOD.Abd3 common bile duct early in disease demonstrates upregulation of genes involved in cholangiocyte injury/morphology and downregulation of immunoregulatory genes. Consistent with this, bone marrow chimera studies show that aberrant Pkhd1 must be expressed in the target tissue (cholangiocytes) and the immune system (bone marrow). Mutations of Pkhd1 produce biliary abnormalities in mice but have not been previously associated with autoimmunity. In this study, we eliminate clinical biliary disease by backcrossing this Pkhd1 mutation onto the C57BL/6 genetic background; thus, the NOD genetic background (which promotes autoimmunity) is essential for disease. We propose that loss of functional Pkhd1 on the NOD background produces early bile duct abnormalities, initiating a break in tolerance that leads to autoimmune cholangitis in NOD.Abd3 congenic mice. This model is important for understanding loss of tolerance to cholangiocytes and is relevant to the pathogenesis of several human cholangiopathies.


Asunto(s)
Enfermedades Autoinmunes/genética , Colangitis/genética , Diabetes Mellitus/genética , Cirrosis Hepática Biliar/genética , Mutación/genética , Receptores de Superficie Celular/genética , Animales , Quimera , Modelos Animales de Enfermedad , Antecedentes Genéticos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Secuencias Repetidas Terminales/genética
13.
J Immunol ; 198(10): 3857-3868, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28363905

RESUMEN

We previously reported that CD137 (encoded by Tnfrsf9) deficiency suppressed type 1 diabetes (T1D) progression in NOD mice. We also demonstrated that soluble CD137 produced by regulatory T cells contributed to their autoimmune-suppressive function in this model. These results suggest that CD137 can either promote or suppress T1D development in NOD mice depending on where it is expressed. In this study, we show that NOD.Tnfrsf9-/- CD8 T cells had significantly reduced diabetogenic capacity, whereas absence of CD137 in non-T and non-B cells had a limited impact on T1D progression. In contrast, NOD.Tnfrsf9-/- CD4 T cells highly promoted T1D development. We further demonstrated that CD137 was important for the accumulation of ß cell-autoreactive CD8 T cells but was dispensable for their activation in pancreatic lymph nodes. The frequency of islet-infiltrating CD8 T cells was reduced in NOD.Tnfrsf9-/- mice in part because of their decreased proliferation. Furthermore, CD137 deficiency did not suppress T1D development in NOD mice expressing the transgenic NY8.3 CD8 TCR. This suggests that increased precursor frequency of ß cell-autoreactive CD8 T cells in NY8.3 mice obviated a role for CD137 in diabetogenesis. Finally, blocking CD137-CD137 ligand interaction significantly delayed T1D onset in NOD mice. Collectively, our results indicate that one important diabetogenic function of CD137 is to promote the expansion and accumulation of ß cell-autoreactive CD8 T cells, and in the absence of CD137 or its interaction with CD137 ligand, T1D progression is suppressed.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/fisiopatología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Ligando 4-1BB/antagonistas & inhibidores , Ligando 4-1BB/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Progresión de la Enfermedad , Células Secretoras de Insulina/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Linfocitos T Reguladores/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/deficiencia , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
14.
J Autoimmun ; 78: 19-28, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28129932

RESUMEN

CXC Chemokine Receptor 3 (CXCR3) is functionally pleiotropic and not only plays an important role in chemotaxis, but also participates in T cell differentiation and may play a critical role in inducing and maintaining immune tolerance. These observations are particularly critical for autoimmune cholangitis in which CXCR3 positive T cells are found around the portal areas of both humans and mouse models of primary biliary cholangitis (PBC). Herein, we investigated the role of CXCR3 in the pathogenesis of autoimmune cholangitis. We have taken advantage of a unique CXCR3 knockout dnTGFßRII mouse to focus on the role of CXCR3, both by direct observation of its influence on the natural course of disease, as well as through adoptive transfer studies into Rag-/- mice. We report herein that not only do CXCR3 deficient mice develop an exacerbation of autoimmune cholangitis associated with an expanded effector memory T cell number, but also selective adoptive transfer of CXCR3 deficient CD8+ T cells induces autoimmune cholangitis. In addition, gene microarray analysis of CXCR3 deficient CD8+ T cells reveal an intense pro-inflammatory profile. Our data suggests that the altered gene profiles induced by CXCR3 deficiency promotes autoimmune cholangitis through pathogenic CD8+ T cells. These data have significance for human PBC and other autoimmune liver diseases in which therapeutic intervention might be directed to chemokines and/or their receptors.


Asunto(s)
Autoinmunidad/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Cirrosis Hepática Biliar/genética , Cirrosis Hepática Biliar/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Receptores CXCR3/deficiencia , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Memoria Inmunológica , Ligandos , Cirrosis Hepática Biliar/metabolismo , Cirrosis Hepática Biliar/patología , Ratones , Ratones Noqueados , Receptores CXCR3/metabolismo
15.
J Autoimmun ; 66: 108-17, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26432598

RESUMEN

There is a significant unmet need in the treatment of primary biliary cirrhosis (PBC) despite significant data on the effector pathways that lead to biliary duct damage. We focused attention on a murine model of PBC, the dominant negative transforming growth factor ß receptor II (Tg) mice. To further define the pathways that lead to biliary pathology in these mice, we developed Tg mice deleted of CD4 cells (CD4(-/-)Tg). Interestingly, these mice developed more severe cholangitis than control Tg mice. These mice, which lack CD4 cells, manifested increased levels of IFN-γ produced by effector CD8 cells. It appears that increased cholangitis is due to the absence of CD4 Treg cells. Based on these data, we parabiosed CD4(-/-)Tg mice with established disease at 8-9 weeks of age with C57BL/6 control mice. Such parabiotic "twins" had a significant reduction in autoimmune cholangitis, even though they had established pathology at the time of surgery. We prepared mixed bone marrow chimera mice constructed from CD4(-/-)Tg and CD8(-/-) mice and not only was cholangitis improved, but a decrease in terminally differentiated CD8(+) T effector cells in the presence of wild type CD4 cells was noted. In conclusion, "correcting" the CD4 T cell subset, even in the presence of pathogenic CD8 T cells, is effective in treating autoimmune cholangitis.


Asunto(s)
Enfermedades Autoinmunes/cirugía , Colangitis/cirugía , Cirrosis Hepática Biliar/cirugía , Parabiosis/métodos , Animales , Enfermedades Autoinmunes/inmunología , Conductos Biliares/inmunología , Conductos Biliares/patología , Antígenos CD4/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Colangitis/inmunología , Modelos Animales de Enfermedad , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Hígado/inmunología , Hígado/patología , Cirrosis Hepática Biliar/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/inmunología
16.
J Autoimmun ; 59: 26-37, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25701076

RESUMEN

CD4(+)Foxp3(+) regulatory T cells (Tregs) play a non-redundant role in control of excessive immune responses, and defects in Tregs have been shown both in patients and murine models of primary biliary cirrhosis (PBC), a progressive autoimmune biliary disease. Herein, we took advantage of a murine model of PBC, the dominant negative transforming growth factor ß receptor II (dnTGFßRII) mice, to assess Treg genetic defects and their functional effects in PBC. By using high-resolution microarrays with verification by PCR and protein expression, we found profound and wide-ranging differences between dnTGFßRII and normal, wild type Tregs. Critical transcription factors were down-regulated including Eos, Ahr, Klf2, Foxp1 in dnTGFßRII Tregs. Functionally, dnTGFßRII Tregs expressed an activated, pro-inflammatory phenotype with upregulation of Ccl5, Granzyme B and IFN-γ. Genetic pathway analysis suggested that the primary effect of loss of TGFß pathway signaling was to down regulate immune regulatory processes, with a secondary upregulation of inflammatory processes. These findings provide new insights into T regulatory genetic defects; aberrations of the identified genes or genetic pathways should be investigated in human PBC Tregs. This approach which takes advantage of biologic pathway analysis illustrates the ability to identify genes/pathways that are affected both independently and dependent on abnormalities in TGFß signaling. Such approaches will become increasingly useful in human autoimmunity.


Asunto(s)
Cirrosis Hepática Biliar/inmunología , Linfocitos T Reguladores/inmunología , Animales , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/genética , Granzimas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Interferón gamma/metabolismo , Cirrosis Hepática Biliar/genética , Ratones , Ratones Mutantes , Análisis por Micromatrices , Mutación/genética , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
J Immunol ; 191(4): 1865-72, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23842751

RESUMEN

We have previously shown that i.m. administration of bacterially expressed murine histidyl-tRNA synthetase (HRS) triggers florid muscle inflammation (relative to appropriate control proteins) in various congenic strains of mice. Because severe disease develops even in the absence of adaptive immune responses to HRS, we sought to identify innate immune signaling components contributing to our model of HRS-induced myositis. In vitro stimulation assays demonstrated HRS-mediated activation of HEK293 cells transfected with either TLR2 or TLR4, revealing an excitatory capacity exceeding that of other bacterially expressed fusion proteins. Corresponding to this apparent functional redundancy of TLR signaling pathways, HRS immunization of B6.TLR2(-/-) and B6.TLR4(-/-) single-knockout mice yielded significant lymphocytic infiltration of muscle tissue comparable to that produced in C57BL/6 wild-type mice. In contrast, concomitant elimination of TLR2 and TLR4 signaling in B6.TLR2(-/-).TLR4(-/-) double-knockout mice markedly reduced the severity of HRS-induced muscle inflammation. Complementary subfragment analysis demonstrated that aa 60-90 of HRS were absolutely required for in vitro as well as in vivo signaling via these MyD88-dependent TLR pathways--effects mediated, in part, through preferential binding of exogenous ligands capable of activating specific TLRs. Collectively, these experiments indicate that multiple MyD88-dependent signaling cascades contribute to this model of HRS-induced myositis, underscoring the antigenic versatility of HRS and confirming the importance of innate immunity in this system.


Asunto(s)
Autoantígenos/toxicidad , Histidina-ARNt Ligasa/toxicidad , Factor 88 de Diferenciación Mieloide/fisiología , Enfermedad Autoinmune Experimental del Sistema Nervioso/fisiopatología , Transducción de Señal/fisiología , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología , Animales , Autoantígenos/química , Autoantígenos/inmunología , Diglicéridos/metabolismo , Endotoxinas/metabolismo , Femenino , Proteína HMGB1/metabolismo , Histidina-ARNt Ligasa/química , Histidina-ARNt Ligasa/inmunología , Inmunidad Innata , Inmunización , Epítopos Inmunodominantes/química , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/toxicidad , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Enfermedad Autoinmune Experimental del Sistema Nervioso/etiología , Oligopéptidos/metabolismo , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Unión Proteica , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/toxicidad , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/efectos de los fármacos , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/efectos de los fármacos
18.
Semin Liver Dis ; 34(3): 285-96, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25057952

RESUMEN

Within the last decade, several mouse models that manifest characteristic features of primary biliary cirrhosis (PBC) with antimitochondrial antibodies (AMAs) and immune-mediated biliary duct pathology have been reported. Here, the authors discuss the current findings on two spontaneous (nonobese diabetic autoimmune biliary disease [NOD.ABD] and dominant negative transforming growth factor-ß receptor II [dnTGFßRII]) and two induced (chemical xenobiotics and microbial immunization) models of PBC. These models exhibit the serological, immunological, and histopathological features of human PBC. From these animal models, it is evident that the etiology of PBC is multifactorial and requires both specific genetic predispositions and environmental insults (either xenobiotic chemicals or microbial), which lead to the breaking of tolerance and eventually liver pathology. Human PBC is likely orchestrated by multiple factors and hence no single model can fully mimic the immunopathophysiology of human PBC. Nevertheless, knowledge gained from these models has greatly advanced our understanding of the major immunological pathways as well as the etiology of PBC.


Asunto(s)
Autoinmunidad , Conductos Biliares Intrahepáticos/inmunología , Modelos Animales de Enfermedad , Cirrosis Hepática Biliar/inmunología , Animales , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/microbiología , Conductos Biliares Intrahepáticos/patología , Biomarcadores/sangre , Escherichia coli/patogenicidad , Humanos , Cirrosis Hepática Biliar/sangre , Cirrosis Hepática Biliar/inducido químicamente , Cirrosis Hepática Biliar/genética , Cirrosis Hepática Biliar/metabolismo , Cirrosis Hepática Biliar/microbiología , Cirrosis Hepática Biliar/patología , Ratones Endogámicos NOD , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Sphingomonadaceae/patogenicidad , Xenobióticos
19.
J Autoimmun ; 50: 123-34, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24556277

RESUMEN

Primary biliary cirrhosis (PBC) is an enigmatic disease mediated by autoimmune destruction of cholangiocytes in hepatic bile ducts. The early immunological events leading to PBC are poorly understood; clinical signs of disease occur very late in the pathological process. We have used our unique murine model of PBC in dominant-negative TGF-ß receptor type II transgenic mice to delineate critical early immunopathological pathways, and previously showed that dnTGFßRII CD8 T cells transfer biliary disease. Herein we report significantly increased numbers of hepatic dnTGFßRII terminally differentiated (KLRG1(+)) CD8 T cells, a CD8 subset previously shown to be enriched in antigen specific cells during hepatic immune response to viral infections. We performed bone marrow chimera studies to assess whether dnTGFßRII CD8 mediated disease was cell intrinsic or extrinsic. Unexpectedly, mixed (dnTGFßRII and B6) bone marrow chimeric (BMC) mice were protected from biliary disease compared to dnTGFßRII single bone marrow chimerics. To define the protective B6 cell subset, we performed adoptive transfer studies, which showed that co-transfer of B6 Tregs prevented dnTGFßRII CD8 T cell mediated cholangitis. Treg mediated disease protection was associated with significantly decreased numbers of hepatic KLRG1(+) CD8 T cells. In contrast, co-transfer of dnTGFßRII Tregs offered no protection, and dnTGFßRII Treg cells were functionally defective in suppressing effector CD8 T cells in vitro compared to wild type B6 Tregs. In vitro cholangiocyte cytotoxicity assays demonstrated significantly increased numbers of cytotoxic hepatic dnTGFßRII KLRG1(+) CD8 cells compared to B6. Protection from disease by B6 Tregs was associated with elimination of hepatic dnTGFßRII CD8 mediated cholangiocyte cytotoxicity. These results emphasize that autoimmune cholangitis requires defects in both the T effector and regulatory compartments, and that an intrinsic T cell effector defect is not sufficient to mediate autoimmune biliary disease in the setting of intact immune regulation. These results have important implications for understanding the early pathogenesis of human PBC.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Colangitis/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Enfermedades Autoinmunes , Médula Ósea/inmunología , Médula Ósea/patología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Linaje de la Célula/inmunología , Quimera/genética , Quimera/inmunología , Colangitis/genética , Colangitis/patología , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Lectinas Tipo C , Cirrosis Hepática Biliar/genética , Cirrosis Hepática Biliar/inmunología , Cirrosis Hepática Biliar/patología , Ratones , Ratones Transgénicos , Receptores Inmunológicos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/trasplante
20.
Hepatology ; 58(3): 1094-104, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23532950

RESUMEN

UNLABELLED: There are several murine models described with features similar to human primary biliary cirrhosis (PBC). Among these models, the one which has the closest serologic features to PBC is a mouse with a T-cell-restricted expression of the dominant negative transforming growth factor ß receptor type II (dnTGFßRII). Our work has demonstrated that CD8(+) T cells from dnTGFßRII mice transfer autoimmune cholangitis to Rag1(-/-) recipients. However, it remained unclear whether the autoimmune cholangitis was secondary to an intrinsic function within CD8(+) T cells or due to the abnormal TGFßR environment within which CD8(+) T cells were generated. To address this mechanistic issue, we used our dnTGFßRII, OT-I/Rag1(-/-) , OT-II/Rag1(-/-) mice and in addition generated OT-I/dnTGFßRII/Rag1(-/-) , and OT-II/dnTGFßRII/Rag1(-/-) mice in which the entire T-cell repertoire was replaced with ovalbumin (OVA)-specific CD8(+) or CD4(+) T cells, respectively. Importantly, neither the parental OT-I/dnTGFßRII/Rag1(-/-) mice and/or OT-II/dnTGFßRII/Rag1(-/-) mice developed cholangitis. However, adoptive transfer demonstrated that only transfer of CD8(+) T cells from dnTGFßRII mice but not CD8(+) T cells from OT-I/Rag1(-/-) mice or from OT-I/dnTGFßRII/Rag1(-/-) mice transferred disease. These data were not secondary to an absence of CD4(+) T cell help since a combination of CD8(+) T cells from OT-I/dnTGFßRII/Rag1(-/-) and CD4(+) T cells from OT II/dnTGFßRII/Rag1(-/-) or CD8(+) T cells from OT-I/dnTGFßRII/Rag1(-/-) with CD4(+) T cells from OT-II/Rag1(-/-) mice failed to transfer disease. CONCLUSION: Defective TGFßRII signaling, in addition to clonal CD8(+) T cells that target biliary cells, are required for induction of autoimmune cholangitis.


Asunto(s)
Enfermedades Autoinmunes/fisiopatología , Linfocitos T CD8-positivos/patología , Colangitis/fisiopatología , Modelos Animales de Enfermedad , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Traslado Adoptivo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Colangitis/inmunología , Colangitis/metabolismo , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Especificidad del Receptor de Antígeno de Linfocitos T/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA