Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Ann Rheum Dis ; 81(8): 1151-1161, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35470161

RESUMEN

OBJECTIVE: Neonatal lupus erythematosus (NLE) may develop after transplacental transfer of maternal autoantibodies with cardiac manifestations (congenital heart block, CHB) including atrioventricular block, atrial and ventricular arrhythmias, and cardiomyopathies. The association with anti-Ro/SSA antibodies is well established, but a recurrence rate of only 12%-16% despite persisting maternal autoantibodies suggests that additional factors are required for CHB development. Here, we identify fetal genetic variants conferring risk of CHB and elucidate their effects on cardiac function. METHODS: A genome-wide association study was performed in families with at least one case of CHB. Gene expression was analysed by microarrays, RNA sequencing and PCR and protein expression by western blot, immunohistochemistry, immunofluorescence and flow cytometry. Calcium regulation and connectivity were analysed in primary cardiomyocytes and cells induced from pleuripotent stem cells. Fetal heart performance was analysed by Doppler/echocardiography. RESULTS: We identified DNAJC6 as a novel fetal susceptibility gene, with decreased cardiac expression of DNAJC6 associated with the disease risk genotype. We further demonstrate that fetal cardiomyocytes deficient in auxilin, the protein encoded by DNAJC6, have abnormal connectivity and Ca2+ homoeostasis in culture, as well as decreased cell surface expression of the Cav1.3 calcium channel. Doppler echocardiography of auxilin-deficient fetal mice revealed cardiac NLE abnormalities in utero, including abnormal heart rhythm with atrial and ventricular ectopias, as well as a prolonged atrioventricular time intervals. CONCLUSIONS: Our study identifies auxilin as the first genetic susceptibility factor in NLE modulating cardiac function, opening new avenues for the development of screening and therapeutic strategies in CHB.


Asunto(s)
Bloqueo Atrioventricular , Auxilinas , Animales , Anticuerpos Antinucleares , Bloqueo Atrioventricular/genética , Autoanticuerpos , Corazón Fetal , Estudio de Asociación del Genoma Completo , Bloqueo Cardíaco/congénito , Lupus Eritematoso Sistémico/congénito , Ratones
2.
Scand J Immunol ; 93(1): e12995, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33188653

RESUMEN

Autoimmune congenital heart block (CHB) may develop in foetuses of women carrying anti-Ro/SSA and La/SSB autoantibodies and is characterized by disruption of signal conduction at the atrioventricular (AV) node, resulting in partial or complete AV block. If not fatal in utero, complete CHB typically requires lifelong cardiac pacing. No treatment has so far been unequivocally demonstrated to prevent or treat autoimmune CHB, and the relatively low incidence (1%-5%) and recurrence (12%-16%) rates of second/third-degree AV block add to the complexity of managing pregnancies in women with anti-Ro/La antibodies. Altogether, a better understanding of events leading to development of autoimmune CHB is needed to improve surveillance and treatment strategies. In the past decade, studies have started to look beyond the role of maternal autoantibodies in disease pathogenesis to assess other contributing factors such as foetal genetics and, more recently, immune responses in foetuses and neonates of anti-Ro/La antibody-positive women. In this review, we provide an update on the epidemiology, clinical presentation and current treatment approaches of autoimmune CHB, summarize the previously proposed pathogenic mechanisms implicating maternal autoantibodies, and discuss the recent findings of type I interferon (IFN) and innate immune activation in foetuses with autoimmune CHB and in neonates of anti-Ro/La antibody-positive mothers, and how these may contribute to autoimmune CHB pathogenesis.


Asunto(s)
Autoinmunidad , Susceptibilidad a Enfermedades , Bloqueo Cardíaco/congénito , Inmunidad Innata , Interferones/metabolismo , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Fibrosis , Bloqueo Cardíaco/diagnóstico , Bloqueo Cardíaco/epidemiología , Bloqueo Cardíaco/etiología , Bloqueo Cardíaco/metabolismo , Humanos , Inmunoterapia , Incidencia , Macrófagos/inmunología , Macrófagos/metabolismo , Recurrencia
3.
Ann Rheum Dis ; 78(5): 696-703, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30808622

RESUMEN

OBJECTIVE: Congenital heart block (CHB) may develop in fetuses of Ro/SSA autoantibody-positive women. Given the rarity of CHB, information on comorbidity and complications later in life is difficult to systematically collect for large groups of patients. We therefore used nation-wide healthcare registers to investigate comorbidity and outcomes in patients with CHB and their siblings. METHODS: Data from patients with CHB (n= 119) and their siblings (n= 128), all born to anti-Ro/SSA-positive mothers, and from matched healthy controls (n= 1,190) and their siblings (n= 1,071), were retrieved from the Swedish National Patient Register. Analyses were performed by Cox proportional hazard modelling. RESULTS: Individuals with CHB had a significantly increased risk of cardiovascular comorbidity, with cardiomyopathy and/or heart failure observed in 20 (16.8%) patients versus 3 (0.3%) controls, yielding a HR of 70.0 (95% CI 20.8 to 235.4), and with a HR for cerebral infarction of 39.9 (95% CI 4.5 to 357.3). Patients with CHB also had a higher risk of infections. Pacemaker treatment was associated with a decreased risk of cerebral infarction but increased risks of cardiomyopathy/heart failure and infection. The risk of systemic connective tissue disorder was also increased in patients with CHB (HR 11.8, 95% CI 4.0 to 11.8), and both patients with CHB and their siblings had an increased risk to develop any of 15 common autoimmune conditions (HR 5.7, 95% CI 2.83 to 11.69 and 3.6, 95% CI 1.7 to 8.0, respectively). CONCLUSIONS: The data indicate an increased risk of several cardiovascular, infectious and autoimmune diseases in patients with CHB, with the latter risk shared by their siblings.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Autoanticuerpos/inmunología , Bloqueo Cardíaco/congénito , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/inmunología , Adolescente , Adulto , Enfermedades Autoinmunes/inmunología , Niño , Preescolar , Comorbilidad , Femenino , Bloqueo Cardíaco/inmunología , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Linaje , Embarazo , Complicaciones del Embarazo/inmunología , Sistema de Registros , Hermanos , Suecia , Adulto Joven
4.
Ann Rheum Dis ; 76(10): 1755-1763, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28760805

RESUMEN

OBJECTIVES: Vaccination of patients with rheumatic disease has been reported to result in lower antibody titres than in healthy individuals. However, studies primarily include patients on immunosuppressive therapy. Here, we investigated the immune response of treatment-naïve patients diagnosed with primary Sjögren's syndrome (pSS) to an H1N1 influenza vaccine. METHODS: Patients with Sjögren's syndrome without immunomodulatory treatment and age-matched and gender-matched healthy controls were immunised with an H1N1 influenza vaccine and monitored for serological and cellular immune responses. Clinical symptoms were monitored with a standardised form. IgG class switch and plasma cell differentiation were induced in vitro in purified naïve B cells of untreated and hydroxychloroquine-treated patients and healthy controls. Gene expression was assessed by NanoString technology. RESULTS: Surprisingly, treatment-naïve patients with Sjögren's syndrome developed higher H1N1 IgG titres of greater avidity than healthy controls on vaccination. Notably, off-target B cells were also triggered resulting in increased anti-EBV and autoantibody titres. Endosomal toll-like receptor activation of naïve B cells in vitro revealed a greater propensity of patient-derived cells to differentiate into plasmablasts and higher production of class switched IgG. The amplified plasma cell differentiation and class switch could be induced in cells from healthy donors by preincubation with type 1 interferon, but was abolished in hydroxychloroquine-treated patients and after in vitro exposure of naïve B cells to chloroquine. CONCLUSIONS: This comprehensive analysis of the immune response in autoimmune patients to exogenous stimulation identifies a mechanistic basis for the B cell hyperactivity in Sjögren's syndrome, and suggests that caution is warranted when considering vaccination in non-treated autoimmune patients.


Asunto(s)
Anticuerpos Antivirales/sangre , Linfocitos B , Citocinas/sangre , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Síndrome de Sjögren/inmunología , Antígenos CD19/análisis , Antirreumáticos/farmacología , Autoanticuerpos/biosíntesis , Autoantígenos/inmunología , Linfocitos B/química , Linfocitos B/fisiología , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Expresión Génica , Antígenos HLA-DR/análisis , Herpesvirus Humano 4/inmunología , Humanos , Hidroxicloroquina/farmacología , Inmunoglobulina D/análisis , Inmunoglobulina G/sangre , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Interleucina-10/farmacología , Activación de Linfocitos , Recuento de Linfocitos , Ribonucleoproteínas/inmunología , Transducción de Señal/genética , Síndrome de Sjögren/genética , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Transcriptoma , Vacunación , Antígeno SS-B
5.
Rheumatology (Oxford) ; 56(10): 1755-1762, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28957562

RESUMEN

Objectives: Congenital heart block (CHB) develops in 1-2% of anti-Ro/SSA-positive pregnancies and has a recurrence rate of 12-20%, which indicates that factors other than maternal autoantibodies are crucial for CHB to occur. Here, we aimed to evaluate the influence of factors previously associated with CHB on the occurrence of milder forms of fetal cardiac conduction disturbances, shown to occur in up to 30% of anti-Ro/SSA-positive pregnancies, and on neonatal outcome in a large cohort of prospectively followed pregnancies. Methods: The association of maternal age, season of the year and history of atrioventricular block (AVB) with the development of fetal Doppler and neonatal ECG conduction disturbances was evaluated in 212 anti-Ro52/SSA-positive singleton pregnancies. Results: Maternal age was significantly higher in AVB II-III pregnancies but was not correlated with fetal AV time intervals in fetuses without signs of AVB II-III. AV time intervals of fetuses surveilled during the winter were significantly longer than those of fetuses surveilled during the summer. Fetal AV time intervals in consecutive pregnancies from the same women were significantly correlated. A history of AVB II-III was associated with significantly longer AV time intervals, and AVB I-III was observed at birth in 38% of babies born after a sibling with abnormal fetal AV conduction. Conclusion: Our study shows that AV time intervals in anti-Ro/SSA antibody-exposed fetuses during the CHB risk period are influenced by the season of the year, and reveals that the recurrence of conduction disturbances in antibody-exposed fetuses is higher than previously reported when milder forms are taken into account.


Asunto(s)
Anticuerpos Antinucleares , Bloqueo Atrioventricular/fisiopatología , Corazón Fetal/fisiopatología , Bloqueo Cardíaco/congénito , Complicaciones Cardiovasculares del Embarazo/inmunología , Estaciones del Año , Adulto , Anticuerpos Antinucleares/sangre , Bloqueo Atrioventricular/congénito , Autoanticuerpos/sangre , Ecocardiografía Doppler/métodos , Femenino , Corazón Fetal/diagnóstico por imagen , Bloqueo Cardíaco/diagnóstico por imagen , Bloqueo Cardíaco/fisiopatología , Humanos , Embarazo , Complicaciones Cardiovasculares del Embarazo/sangre , Complicaciones Cardiovasculares del Embarazo/diagnóstico por imagen , Resultado del Embarazo , Estudios Prospectivos , Ribonucleoproteínas/inmunología , Factores de Riesgo , Ultrasonografía Prenatal/métodos
6.
Curr Opin Rheumatol ; 27(5): 468-75, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26108063

RESUMEN

PURPOSE OF REVIEW: The purpose of this review is to give an update on the understanding of the immune responses involved in the pathogenesis of primary Sjögren's syndrome (pSS), and to highlight recent findings on the underlying molecular and cellular mechanisms at play. RECENT FINDINGS: In recent years, genetic studies have confirmed the importance of aberrant type I interferon (IFN) and B cell responses in pSS and highlighted critical pathways involved in disease pathogenesis. In particular, the formation of ectopic lymphoid structures has emerged as an important factor in the establishment of chronic autoimmune responses in target organs. Interestingly, recent studies on viral infection in the context of pSS, as well as findings on the contribution of salivary gland epithelial cells in local immune responses, offer further clues to understand pSS etiology and its target organ specificity. Finally, new evidence brings T cells and natural killer cells under renewed attention as possible important contributors to pSS pathogenesis. SUMMARY: Progress made during the last few years on the pathogenesis of pSS has been mirrored by clinical trials directed at inhibiting cytokines, B, or T cell responses. Future efforts should focus on identifying additional pSS specific targets and developing methods to help choose optimal therapeutic strategies for the individual patient.


Asunto(s)
Síndrome de Sjögren/inmunología , Inmunidad Adaptativa , Linfocitos B/inmunología , Humanos , Interferón Tipo I/inmunología , Células Asesinas Naturales/inmunología , Glándulas Salivales/inmunología , Síndrome de Sjögren/virología , Linfocitos T/inmunología
7.
J Immunol ; 191(7): 3753-63, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23975864

RESUMEN

Tripartite-motif 21 (TRIM21) is an E3 ubiquitin ligase that regulates innate immune responses by ubiquitinating IFN regulatory factors (IRFs). TRIM21 is mainly found in hematopoietic cells in which its expression is induced by IFNs during viral. infections and in systemic autoimmune diseases such as systemic lupus erythematosus and Sjögren's syndrome. However, the exact molecular mechanism by which the expression of the Trim21 gene is regulated is unknown. In this study, we demonstrate that IFNs induce Trim21 expression in immune cells via IRFs and that IFN-α and IFN-ß are the most potent inducers of Trim21. A functional IFN-stimulated response element but no conserved IFN-γ-activated site was detected in the promoter of Trim21. IRF1 and IRF2 strongly induced Trim21 expression in an IFN-stimulated response element-dependent manner, whereas IRF4 and IRF8 strongly repressed the IRF1-mediated induction of Trim21. Consistent with this observation, baseline expression of Trim21 was elevated in Irf4(-/-) cells. TRIM21, IRF1, and IRF2 expression was increased in PBMCs from patients with Sjögren's syndrome compared with healthy controls. In contrast, IRF4 and IRF8 expression was not increased in PBMCs from patients. The IFN-γ-mediated induction of Trim21 was completely abolished by inhibiting protein synthesis with cycloheximide, and Trim21 expression could not be induced by IFN-γ in Irf1(-/-) cells, demonstrating that IFN-γ induces Trim21 indirectly via IRF1 and not directly via STAT1 activation. Our data demonstrate that multiple IRFs tightly regulate expression of Trim21 in immune cells, suggesting that a well-controlled expression of the E3 ligase TRIM21 is important for regulation of immune responses.


Asunto(s)
Regulación de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Ribonucleoproteínas/genética , Animales , Secuencia de Bases , Línea Celular , Secuencia Conservada , Orden Génico , Humanos , Factor 1 Regulador del Interferón/metabolismo , Factor 2 Regulador del Interferón/metabolismo , Interferón-alfa/farmacología , Interferón beta/farmacología , Interferón gamma/farmacología , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Elementos de Respuesta , Factor de Transcripción STAT1/metabolismo , Síndrome de Sjögren/genética , Síndrome de Sjögren/metabolismo
8.
Exp Cell Res ; 325(1): 2-9, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24434353

RESUMEN

Autoantibody-associated congenital heart block (CHB) is a passively acquired autoimmune condition associated with maternal anti-Ro/SSA antibodies and primarily affecting electric signal conduction at the atrioventricular node in the fetal heart. CHB occurs in 1-2% of anti-Ro/SSA antibody-positive pregancies and has a recurrence rate of 12-20% in a subsequent pregnancy. Despite the long-recognized association between maternal anti-Ro/SSA autoantibodies and CHB, the molecular mechanisms underlying CHB pathogenesis are not fully understood, but several targets for the maternal autoantibodies in the fetal heart have been suggested. Recent studies also indicate that fetal susceptibility genes determine whether an autoantibody-exposed fetus will develop CHB or not, and begin to identify such genes. In this article, we review the different lines of investigation undertaken to elucidate the molecular pathways involved in CHB development and reflect on the hypotheses put forward to explain CHB pathogenesis as well as on the questions left unanswered and that should guide future studies.


Asunto(s)
Bloqueo Cardíaco/congénito , Animales , Apoptosis , Autoanticuerpos/metabolismo , Autoantígenos/inmunología , Bloqueo Cardíaco/inmunología , Humanos , Inflamación , Miocardio/inmunología , Miocardio/patología , ARN Citoplasmático Pequeño/inmunología , Ribonucleoproteínas/inmunología
9.
Acta Paediatr ; 103(3): 275-81, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24164657

RESUMEN

AIM: To investigate the correlation between maternal autoantibodies and age at diagnosis of isolated complete atrioventricular (AV) block (CAVB) and to study signs of late progression of foetal immune-mediated insults in cases of postnatally diagnosed CAVB. METHODS: Patients with CAVB (n = 190) identified in a population-based manner were included. Maternal autoantibody profile was correlated with age at CAVB diagnosis. A structured review of medical records was performed if a late CAVB diagnosis (>27 days post-partum) was associated with a sero-positive mother. RESULTS: Maternal Ro/La autoantibodies were observed in 88% of cases with a congenital diagnosis. Thirteen cases with a sero-positive mother and late CAVB diagnosis were found (age-range: 4 months-43 years). In two cases, CAVB was diagnosed in conjunction with infections, one case had a family history of cardiomyopathy and two cases had nontypical clinical presentations, indicating alternative pathogenetic mechanisms. In the remaining eight cases, no likely factors inducing CAVB, other than maternal autoantibodies, could be identified. CONCLUSION: Our observations support the hypothesis that late progression to CAVB can be the result of an immune-mediated pathogenetic mechanism during foetal life. An autoantibody-associated diagnosis after the neonatal period is therefore possible, and testing of maternal serology at the time of diagnosis is recommended.


Asunto(s)
Bloqueo Atrioventricular/congénito , Bloqueo Atrioventricular/inmunología , Autoanticuerpos/sangre , Adolescente , Adulto , Autoanticuerpos/biosíntesis , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Lactante , Masculino , Embarazo/sangre , Adulto Joven
10.
Eur J Immunol ; 42(9): 2274-84, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22949326

RESUMEN

Systemic autoimmune diseases such as systemic lupus erythematosus are type I IFN-driven diseases with exaggerated B-cell responses and autoantibody production. Th17 cells, a T-helper-cell subset with high inflammatory capacity, was initially discovered and characterized in the context of experimental autoimmune encephalomyelitis - an animal model of multiple sclerosis. There is now emerging evidence that Th17 cells, and more generally IL-17 and IL-17-producing cells, may play a role in the pathogenesis of type I IFN-driven systemic autoimmune diseases such as lupus. Here, we review the different studies suggesting a role for IL-17 and IL-17-producing cells in systemic autoimmune diseases, both in humans and in animal models, and we consider the possible mechanisms by which these cells may contribute to disease. We also discuss the hypothesis that type I IFN and IL-17 act in concert to sustain and amplify autoimmune and inflammatory responses, making them a dangerous combination involved in the pathogenesis of systemic autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Interferón Tipo I/inmunología , Interleucina-17/inmunología , Células Th17/inmunología , Animales , Humanos
11.
J Biol Chem ; 286(42): 36478-91, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21862588

RESUMEN

Ro52 (TRIM21) is an E3 ligase of the tripartite motif family that negatively regulates proinflammatory cytokine production by ubiquitinating transcription factors of the interferon regulatory factor family. Autoantibodies to Ro52 are present in patients with lupus and Sjögren's syndrome, but it is not known if these autoantibodies affect the function of Ro52. To address this question, the requirements for Ro52 E3 ligase activity were first analyzed in detail. Scanning a panel of E2 ubiquitin-conjugating enzymes, we found that UBE2D1-4 and UBE2E1-2 supported the E3 ligase activity of Ro52 and that the E3 ligase activity of Ro52 was dependent on its RING domain. We also found that the N-terminal extensions in the class III E2 enzymes affected their interaction with Ro52. Although the N-terminal extension in UBE2E3 made this E2 enzyme unable to function together with Ro52, the N-terminal extensions in UBE2E1 and UBE2E2 allowed for a functional interaction with Ro52. Anti-Ro52-positive patient sera and affinity-purified anti-RING domain autoantibodies inhibited the E3 activity of Ro52 in ubiquitination assays. Using NMR, limited proteolysis, ELISA, and Ro52 mutants, we mapped the interactions between Ro52, UBE2E1, and anti-Ro52 autoantibodies. We found that anti-Ro52 autoantibodies inhibited the E3 ligase activity of Ro52 by sterically blocking the E2/E3 interaction between Ro52 and UBE2E1. Our data suggest that anti-Ro52 autoantibodies binding the RING domain of Ro52 may be actively involved in the pathogenesis of rheumatic autoimmune disease by inhibiting Ro52-mediated ubiquitination.


Asunto(s)
Autoanticuerpos/inmunología , Ribonucleoproteínas/antagonistas & inhibidores , Ribonucleoproteínas/inmunología , Síndrome de Sjögren/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación/inmunología , Autoanticuerpos/farmacología , Línea Celular , Humanos , Estructura Terciaria de Proteína , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Síndrome de Sjögren/enzimología , Síndrome de Sjögren/genética , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
12.
Ann Rheum Dis ; 71(3): 448-54, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22084395

RESUMEN

BACKGROUND: Congenital heart block (CHB) may develop in fetuses of women with anti-Ro/La autoantibodies following placental transfer of maternal autoantibodies and disruption of the fetal atrioventricular (AV) conduction system. Animal models of CHB currently rely on immunisation or transfer of anti-Ro/La antibodies purified from mothers of children with CHB, which does not allow precise identification of the disease-inducing antibody specificity. OBJECTIVE: To determine the ability of different anti-Ro52 monoclonal antibodies to induce cardiac electrophysiological abnormalities in vivo and affect the calcium homoeostasis of cardiomyocytes in vitro. METHODS: Monoclonal antibodies recognising different domains of Ro52 were generated and injected into pregnant rats, and ECG was recorded on newborn pups. Cultures of rat neonatal cardiomyocytes were established to assess the effect of the different anti-Ro52 monoclonal antibodies on calcium homoeostasis. RESULTS: First-degree AV block and bradycardia developed after maternal transfer of antibodies specific for amino acids 200-239 of Ro52 (p200), while pups exposed to antibodies targeting N- or C-terminal epitopes of Ro52 did not show any electrocardiogram abnormalities. Addition of an anti-p200 antibody to cultured cardiomyocytes induced calcium dyshomoeostasis in a time- and dose-dependent manner, while addition of other Ro52 antibodies had no effect. CONCLUSION: These data for the first time show unambiguously that antibodies specific for amino acids 200-239 of Ro52 can induce cardiac conduction defects in the absence of other autoantibodies, and may therefore be the main initiators of cardiac pathology in the pool of anti-Ro52 antibodies in mothers of children with CHB.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Bloqueo Atrioventricular/congénito , Ribonucleoproteínas/inmunología , Animales , Animales Recién Nacidos , Especificidad de Anticuerpos/inmunología , Bloqueo Atrioventricular/inmunología , Autoanticuerpos/inmunología , Calcio/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Electrocardiografía , Epítopos/inmunología , Femenino , Homeostasis/inmunología , Intercambio Materno-Fetal/inmunología , Miocitos Cardíacos/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inmunología , Ratas
13.
Ann Rheum Dis ; 71(3): 334-40, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21953338

RESUMEN

OBJECTIVE: Congenital heart block may develop in the fetuses of Ro/SSA-positive and La/SSB-positive mothers. Recurrence rates of only 10-20% despite persisting maternal antibodies indicate that additional factors are critical for the establishment of heart block. The authors investigated the influence of other maternal and fetal factors on heart block development in a Swedish population-based cohort. METHODS: The influence of fetal gender, maternal age, parity and time of birth on heart block development was analysed in 145 families, including Ro/La-positive (n=190) and Ro/La-negative (n=165) pregnancies. RESULTS: There was a recurrence rate of 12.1% in Ro/La-positive women, and no recurrence in Ro/La-negative women. Fetal gender and parity did not influence the development of heart block in either group. Maternal age in Ro/La-positive pregnancies with a child affected by heart block was, however, significantly higher than in pregnancies resulting in babies without heart block (p<0.05).Seasonal timing of pregnancy influenced the outcome. Gestational susceptibility weeks 18-24 occurring during January-March correlated with a higher proportion of children with heart block and lower vitamin D levels during the same period in a representative sample of Swedish women and a corresponding higher proportion of children with heart block born in the summer (p<0.02). Maternal age or seasonal timing of pregnancy did not affect the outcome in Ro/La-negative pregnancies. CONCLUSION: This study identifies maternal age and seasonal timing of pregnancy as novel risk factors for heart block development in children of Ro/La-positive women. These observations may be useful for counselling when pregnancy is considered.


Asunto(s)
Anticuerpos Antinucleares/sangre , Bloqueo Cardíaco/congénito , Edad Materna , Estaciones del Año , Adolescente , Adulto , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/inmunología , Orden de Nacimiento , Niño , Preescolar , Composición Familiar , Femenino , Bloqueo Cardíaco/epidemiología , Bloqueo Cardíaco/inmunología , Humanos , Lactante , Recién Nacido , Paridad , Embarazo , Complicaciones del Embarazo/epidemiología , Complicaciones del Embarazo/inmunología , Efectos Tardíos de la Exposición Prenatal , Recurrencia , Factores de Riesgo , Factores Sexuales , Suecia/epidemiología , Vitamina D/sangre , Adulto Joven
14.
J Immunol ; 185(6): 3574-82, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20696861

RESUMEN

Congenital heart block develops in fetuses of anti-Ro52 Ab-positive women. A recurrence rate of 20%, despite the persistence of maternal autoantibodies, indicates that there are additional, yet unidentified, factors critical for development of congenital heart block. In this study, we demonstrate that besides the maternal MHC controlling Ab specificity, fetal MHC-encoded genes influence fetal susceptibility to congenital heart block. Using MHC congenic rat strains, we show that heart block develops in rat pups of three strains carrying MHC haplotype RT1(av1) (DA, PVG.AV1, and LEW.AV1) after maternal Ro52 immunization, but not in LEW rats (RT1(l)). Different anti-Ro52 Ab fine specificities were generated in RT1(av1) versus RT1(l) animals. Maternal and fetal influence was determined in an F(2) cross between LEW.AV1 and LEW strains, which revealed higher susceptibility in RT1(l) than RT1(av1) pups once pathogenic Ro52 Abs were present. This was further confirmed in that RT1(l) pups more frequently developed heart block than RT1(av1) pups after passive transfer of RT1(av1) anti-Ro52 sera. Our findings show that generation of pathogenic Ro52 Abs is restricted by maternal MHC, whereas the fetal MHC locus regulates susceptibility and determines the fetal disease outcome in anti-Ro52-positive pregnancies.


Asunto(s)
Bloqueo Atrioventricular/genética , Bloqueo Atrioventricular/inmunología , Autoanticuerpos/biosíntesis , Predisposición Genética a la Enfermedad , Antígenos de Histocompatibilidad/genética , Intercambio Materno-Fetal/inmunología , Ribonucleoproteínas/inmunología , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos/genética , Bloqueo Atrioventricular/congénito , Línea Celular , Modelos Animales de Enfermedad , Femenino , Antígenos de Histocompatibilidad/inmunología , Intercambio Materno-Fetal/genética , Datos de Secuencia Molecular , Embarazo , Ratas , Ratas Endogámicas Lew , Ribonucleoproteínas/administración & dosificación
15.
Arthritis Res Ther ; 14(2): 208, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22546326

RESUMEN

During pregnancy in autoimmune conditions, maternal autoantibodies are transported across the placenta and may affect the developing fetus. Congenital heart block (CHB) is known to associate with the presence of anti-Ro/SSA and anti-La/SSB antibodies in the mother and is characterized by a block in signal conduction at the atrioventricular (AV) node. The mortality rate of affected infants is 15% to 30%, and most live-born children require lifelong pacemaker implantation. Despite a well-recognized association with maternal anti-Ro/La antibodies, CHB develops in only 1% to 2% of anti-Ro-positive pregnancies, indicating that other factors are important for establishment of the block. The molecular mechanisms leading to complete AV block are still unclear, and the existing hypotheses fail to explain all aspects of CHB in one comprehensive model. In this review, we discuss the different specificities of maternal autoantibodies that have been implicated in CHB as well as the molecular mechanisms that have been suggested to operate, focusing on the evidence supporting a direct pathogenic role of maternal antibodies. Autoantibodies targeting the 52-kDa component of the Ro antigen remain the antibodies most closely associated with CHB. In vitro experiments and animal models of CHB also point to a major role for anti-Ro52 antibodies in CHB pathogenesis and suggest that these antibodies may directly affect calcium regulation in the fetal heart, leading to disturbances in signal conduction or electrogenesis or both. In addition, maternal antibody deposits are found in the heart of fetuses dying of CHB and are thought to contribute to an inflammatory reaction that eventually induces fibrosis and calcification of the AV node, leading to a complete block. Considering that CHB has a recurrence rate of 12% to 20% despite persisting maternal autoantibodies, it has long been clear that maternal autoantibodies are not sufficient for the establishment of a complete CHB, and efforts have been made to identify additional risk factors for this disorder. Therefore, recent studies looking at the influence of genetic and environmental factors will also be discussed.


Asunto(s)
Autoanticuerpos/fisiología , Bloqueo Cardíaco/congénito , Intercambio Materno-Fetal/inmunología , Animales , Autoanticuerpos/biosíntesis , Femenino , Bloqueo Cardíaco/sangre , Bloqueo Cardíaco/etiología , Bloqueo Cardíaco/inmunología , Humanos , Embarazo
16.
J Exp Med ; 206(8): 1661-71, 2009 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-19635858

RESUMEN

Ro52/Trim21 is targeted as an autoantigen in systemic lupus erythematosus and Sjögren's syndrome. Polymorphisms in the Ro52 gene have been linked to these autoimmune conditions, but the molecular mechanism by which Ro52 may promote development of systemic autoimmune diseases has not been explored. To address this issue, we generated Ro52-null mice (Ro52(-/-)), which appear phenotypically normal if left unmanipulated. However, Ro52(-/-) mice develop severe dermatitis extending from the site of tissue injury induced by ear tags. The affected mice further develop several signs of systemic lupus with hypergammaglobulinemia, autoantibodies to DNA, proteinuria, and kidney pathology. Ro52, which was recently identified as an E3 ligase, mediates ubiquitination of several members of the interferon regulatory factor (IRF) family, and the Ro52-deficient mice have an enhanced production of proinflammatory cytokines that are regulated by the IRF transcription factors, including cytokines involved in the Th17 pathway (interleukin [IL] 6, IL-12/IL-23p40, and IL-17). Loss of IL-23/IL-17 by genetic deletion of IL-23/p19 in the Ro52(-/-) mice conferred protection from skin disease and systemic autoimmunity. These data reveal that the lupus-associated Ro52 protein is an important negative regulator of proinflammatory cytokine production, and they provide a mechanism by which a defective Ro52 function can lead to tissue inflammation and systemic autoimmunity through the IL-23-Th17 pathway.


Asunto(s)
Enfermedades Autoinmunes/etiología , Inflamación/etiología , Interleucina-23/metabolismo , Ribonucleoproteínas/deficiencia , Animales , Enfermedades Autoinmunes/patología , Secuencia de Bases , Citocinas/biosíntesis , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Retroalimentación Fisiológica , Humanos , Inflamación/patología , Mediadores de Inflamación/metabolismo , Interferones/farmacología , Interleucina-23/deficiencia , Interleucina-23/genética , Lupus Eritematoso Sistémico/etiología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ribonucleoproteínas/genética , Subgrupos de Linfocitos T/clasificación , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/clasificación , Linfocitos T Colaboradores-Inductores/inmunología
17.
J Clin Immunol ; 28(3): 220-31, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18071879

RESUMEN

Interferon-alpha (IFN-alpha) has been implicated in the pathogenesis of Sjögren's syndrome and systemic lupus erythematosus. Ro52, which was recently identified as an E3 ligase with anti-proliferative and pro-apoptotic properties, is a major autoantigen targeted in both these conditions. Microarray analyses have indicated up-regulation of Ro52 by IFN-alpha, and the objective of the present study was to address the potential link between IFN-alpha and Ro52. To investigate the influence of IFN-alpha on Ro52 protein levels and cellular localization, we generated a panel of monoclonal antibodies to different domains of Ro52. These novel monoclonal antibodies were characterized by immunoprecipitation, Western blot, and enzyme-linked immunosorbent assay using cell lysates, recombinant Ro52 protein, and synthetic peptides. Ro52 was up-regulated in HeLa cells and human B cells at the messenger RNA and protein levels in response to IFN-alpha stimulation as detected by reverse transcriptase polymerase chain reaction and Western blot. After up-regulation, Ro52 translocated from the cytoplasm to the nucleus. The nuclear translocation of Ro52 was observed after staining with generated monoclonal antibodies specific for both the RING, coiled-coil, and B30.2 domains of Ro52 and the nuclear translocation of Ro52 preceded IFN-alpha-induced apoptotic cell death detected by caspase-3 and TUNEL staining in the treated cultures. In conclusion, our data show that IFN-alpha first induces up-regulation of Ro52 protein and then prompts translocation of the up-regulated Ro52 protein in to the nucleus. The translocation precedes apoptosis of the IFN-alpha exposed cells, suggesting a role for Ro52 in mediating the anti-proliferative or pro-apoptotic effects of the autoimmune-related cytokine IFN-alpha.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Autoantígenos/metabolismo , Interferón-alfa/fisiología , Ribonucleoproteínas/inmunología , Ribonucleoproteínas/metabolismo , Regulación hacia Arriba/inmunología , Transporte Activo de Núcleo Celular/inmunología , Animales , Especificidad de Anticuerpos , Apoptosis/inmunología , Línea Celular Tumoral , Núcleo Celular/enzimología , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Proliferación Celular , Células HeLa , Humanos , Ratones , Ratones Endogámicos , Ribonucleoproteínas/fisiología , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA