Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-33172961

RESUMEN

OBJECTIVE: To determine whether maternofetal transfer of NMDA receptor (NMDAR) antibodies has pathogenic effects on the fetus and offspring, we developed a model of placental transfer of antibodies. METHODS: Pregnant C57BL/6J mice were administered via tail vein patients' or controls' immunoglobulin G (IgG) on days 14-16 of gestation, when the placenta is able to transport IgG and the immature fetal blood-brain barrier is less restrictive to IgG crossing. Immunohistochemical and DiOlistic (gene gun delivery of fluorescent dye) staining, confocal microscopy, standardized developmental and behavioral tasks, and hippocampal long-term potentiation were used to determine the antibody effects. RESULTS: In brains of fetuses, patients' IgG, but not controls' IgG, bound to NMDAR, causing a decrease in NMDAR clusters and cortical plate thickness. No increase in neonatal mortality was observed, but offspring exposed in utero to patients' IgG had reduced levels of cell-surface and synaptic NMDAR, increased dendritic arborization, decreased density of mature (mushroom-shaped) spines, microglial activation, and thinning of brain cortical layers II-IV with cellular compaction. These animals also had a delay in innate reflexes and eye opening and during follow-up showed depressive-like behavior, deficits in nest building, poor motor coordination, and impaired social-spatial memory and hippocampal plasticity. Remarkably, all these paradigms progressively improved (becoming similar to those of controls) during follow-up until adulthood. CONCLUSIONS: In this model, placental transfer of patients' NMDAR antibodies caused severe but reversible synaptic and neurodevelopmental alterations. Reversible antibody effects may contribute to the infrequent and limited number of complications described in children of patients who develop anti-NMDAR encephalitis during pregnancy.


Asunto(s)
Autoanticuerpos/toxicidad , Encéfalo/patología , Efectos Tardíos de la Exposición Prenatal , Animales , Conducta Animal , Femenino , Humanos , Inmunoglobulina G , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos C57BL , Placenta , Embarazo , Complicaciones del Embarazo
2.
Acta Biochim Biophys Sin (Shanghai) ; 52(12): 1373-1381, 2020 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-33231607

RESUMEN

Exposure to adverse factors in utero may lead to adaptive changes in cardiac structure and metabolism, which increases the risk of chronic cardiovascular disease later in life. Studies showed that the angiotensin II type 1 receptor autoantibodies (AT1-AAs) are able to cross the placenta into the circulation of pregnant rodents' embryo, which adversely affects embryogenesis. However, the effects of AT1-AA exposure on the fetal heart in utero are still unknown. In this study, we investigated whether intrauterine AT1-AA exposure has adverse effects on fetal heart structure, function and metabolism. AT1-AA-positive pregnant mouse models were successfully established by passive immunity, evidenced by increased AT1-AA content. Morphological and ultrasonic results showed that the fetal mice on embryonic day 18 (E18) of AT1-AA group have loose and disordered myocardial structure, and decreased left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS), compared with control groups. The myocardium of AT1-AA group fetal mice on E18 exhibited increased expression of the key molecules in the glycolytic pathway, pyruvate and lactic acid content and ATP production, suggesting that the glycolysis rate was enhanced. Furthermore, the enhanced effect of glycolysis caused by AT1-AA is mainly through the PPARß/δ pathway. These data confirmed that fetus exposure to AT1-AA in utero developed left ventricular dysfunction, myocardial structural arrangement disorders, and enhanced glycolysis on E18. Our results support AT1-AA being a potentially harmful factor for cardiovascular disease in fetal mice.


Asunto(s)
Autoanticuerpos/toxicidad , Cardiomiopatías/etiología , Feto/inmunología , Feto/fisiopatología , Glucólisis/inmunología , Receptor de Angiotensina Tipo 1/inmunología , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Cardiomiopatías/patología , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C , PPAR gamma/metabolismo , PPAR-beta/metabolismo , Placenta/fisiología , Embarazo , Segundo Trimestre del Embarazo , Volumen Sistólico/inmunología , Función Ventricular Izquierda/inmunología
3.
J Neuroinflammation ; 17(1): 31, 2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31973738

RESUMEN

OBJECTIVE: Autoantibodies against ribosomal P proteins (anti-P antibodies) are strongly associated with the neuropsychiatric manifestations of systemic lupus erythematosus (NPSLE). The present study was designed to assess whether anti-P antibodies can induce abnormal brain electrical activities in mice and investigate the potential cytopathological mechanism. METHODS: Affinity-purified human anti-ribosomal P antibodies were injected intravenously into mice after blood-brain barrier (BBB) disruption. The auditory steady-state response (ASSR) was evaluated based on electroencephalography (EEG) signals in response to 40-Hz click-train stimuli, which were recorded from electrodes implanted in the skull of mice. Immunofluorescence staining was used to examine the morphology and density of neurons and glia in the hippocampus and cortex. The presence of apoptosis in the brain tissues was studied using the TUNEL assay. A PLX3397 diet was used to selectively eliminate microglia from the brains of mice. RESULTS: Circulating anti-P antibodies caused an enhancement of the ASSR and the activation of microglia through the disrupted BBB, while no obvious neural apoptosis was observed. In contrast, when microglia were depleted, anti-P antibodies induced a serious reduction in the ASSR and neural apoptosis. CONCLUSION: Our study indicates that anti-P antibodies can directly induce the dysfunction of auditory-evoked potentials in the brain and that microglia are involved in the protection of neural activity after the invasion of anti-P antibodies, which could have important implications for NPSLE.


Asunto(s)
Autoanticuerpos/toxicidad , Potenciales Evocados Auditivos/efectos de los fármacos , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Microglía/inmunología , Proteínas Ribosómicas/inmunología , Animales , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Proc Natl Acad Sci U S A ; 116(44): 22246-22251, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31611368

RESUMEN

Immune checkpoint (IC) therapy provides substantial benefits to cancer patients but can also cause distinctive toxicities termed immune-related adverse events (irAEs). Biomarkers to predict toxicities will be necessary to improve management of patients receiving IC therapy. We relied on serological analysis of recombinant cDNA expression libraries to evaluate plasma samples from patients treated with IC therapy and identified autoantibodies, both in pretreatment and on-treatment samples prior to the development of irAEs, which correlate with the development of immune-related hypophysitis (anti-GNAL and anti-ITM2B autoantibodies) and pneumonitis (anti-CD74 autoantibody). We developed an enzyme-linked immunosorbent assay and tested additional patient samples to confirm our initial findings. Collectively, our data suggest that autoantibodies may correlate with irAEs related to IC therapy, and specific autoantibodies may be detected early for the management of irAEs.


Asunto(s)
Autoanticuerpos/inmunología , Hipofisitis Autoinmune/etiología , Inmunoterapia/efectos adversos , Neumonía/etiología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Anciano , Autoanticuerpos/sangre , Autoanticuerpos/toxicidad , Hipofisitis Autoinmune/diagnóstico , Hipofisitis Autoinmune/inmunología , Biomarcadores/sangre , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/inmunología , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/terapia , Neumonía/inmunología
5.
Ann Neurol ; 86(5): 656-670, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31325344

RESUMEN

OBJECTIVE: Maternal autoantibodies are a risk factor for impaired brain development in offspring. Antibodies (ABs) against the NR1 (GluN1) subunit of the N-methyl-d-aspartate receptor (NMDAR) are among the most frequently diagnosed anti-neuronal surface ABs, yet little is known about effects on fetal development during pregnancy. METHODS: We established a murine model of in utero exposure to human recombinant NR1 and isotype-matched nonreactive control ABs. Pregnant C57BL/6J mice were intraperitoneally injected on embryonic days 13 and 17 each with 240µg of human monoclonal ABs. Offspring were investigated for acute and chronic effects on NMDAR function, brain development, and behavior. RESULTS: Transferred NR1 ABs enriched in the fetus and bound to synaptic structures in the fetal brain. Density of NMDAR was considerably reduced (up to -49.2%) and electrophysiological properties were altered, reflected by decreased amplitudes of spontaneous excitatory postsynaptic currents in young neonates (-34.4%). NR1 AB-treated animals displayed increased early postnatal mortality (+27.2%), impaired neurodevelopmental reflexes, altered blood pH, and reduced bodyweight. During adolescence and adulthood, animals showed hyperactivity (+27.8% median activity over 14 days), lower anxiety, and impaired sensorimotor gating. NR1 ABs caused long-lasting neuropathological effects also in aged mice (10 months), such as reduced volumes of cerebellum, midbrain, and brainstem. INTERPRETATION: The data collectively support a model in which asymptomatic mothers can harbor low-level pathogenic human NR1 ABs that are diaplacentally transferred, causing neurotoxic effects on neonatal development. Thus, AB-mediated network changes may represent a potentially treatable neurodevelopmental congenital brain disorder contributing to lifelong neuropsychiatric morbidity in affected children. ANN NEUROL 2019;86:656-670.


Asunto(s)
Autoanticuerpos/toxicidad , Encéfalo/patología , Efectos Tardíos de la Exposición Prenatal , Receptores de N-Metil-D-Aspartato/inmunología , Animales , Autoantígenos/inmunología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Discapacidades del Desarrollo/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Embarazo , Receptores de N-Metil-D-Aspartato/metabolismo
6.
Int J Mol Sci ; 20(10)2019 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-31137749

RESUMEN

Studies have suggested an involvement of the immune system in glaucoma. Hence, a rat experimental autoimmune glaucoma model (EAG) was developed to investigate the role of the immune response. Here, we transferred this model into mice. Either 0.8 mg/mL of the optic nerve antigen homogenate (ONA; ONA 0.8) or 1.0 mg/mL ONA (ONA 1.0) were injected in 129/Sv mice. Controls received sodium chloride. Before and 6 weeks after immunization, the intraocular pressure (IOP) was measured. At 6 weeks, retinal neurons, glia cells, and synapses were analyzed via immunohistology and quantitative real-time PCR (RT-qPCR). Additionally, optic nerves were examined. The IOP stayed in the normal physiological range throughout the study (p > 0.05). A significant reduction of retinal ganglion cells (RGCs) was noted in both immunized groups (p < 0.001). Remodeling of glutamatergic and GABAergic synapses was seen in ONA 1.0 retinas. Furthermore, both ONA groups revealed optic nerve degeneration and macrogliosis (all: p < 0.001). An increase of activated microglia was noted in ONA retinas and optic nerves (p < 0.05). Both ONA concentrations led to RGC loss and optic nerve degeneration. Therefore, the EAG model was successfully transferred from rats to mice. In further studies, transgenic knockout mice can be used to investigate the pathomechanisms of glaucoma more precisely.


Asunto(s)
Autoanticuerpos/toxicidad , Enfermedades Autoinmunes del Sistema Nervioso/patología , Glaucoma/patología , Retina/patología , Animales , Autoanticuerpos/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/fisiopatología , Modelos Animales de Enfermedad , Glaucoma/inmunología , Glaucoma/fisiopatología , Presión Intraocular , Ratones , Nervio Óptico/inmunología , Nervio Óptico/patología , Retina/inmunología , Sinapsis/patología
7.
Front Immunol ; 9: 1935, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30233569

RESUMEN

Pemphigus vulgaris (PV) is a potentially life-threatening autoimmune blistering disease which is associated with autoantibodies directed against two desmosomal proteins, desmoglein (Dsg) 3 and 1. Treatment of PV is rather challenging and relies on the long-term use of systemic corticosteroids and additional immunosuppressants. More recently, autoantibody-depleting therapies such as rituximab, high-dose intravenous immunoglobulins, and immunoadsorption were shown to be valuable treatment options in PV. Specific removal of pathogenic autoantibodies would further increase efficacy and usability of immunoadsorption. Here, we tested the capacity of our recently developed prototypic Dsg1- and Dsg3-specific adsorbers to remove circulating pathogenic autoantibodies from three different PV patients. The pathogenic potential of the Dsg3/1-depleted IgG fractions and the anti-Dsg3-specific IgG was explored in two different in vitro assays based on cultured human keratinocytes, the desmosome degradation assay and the dispase-based dissociation assay. In addition, the neonatal mouse model of PV was used. In both in vitro assays, no difference between the pathogenic effect of total PV IgG and anti-Dsg3-specific IgG was seen, while Dsg3/1-depleted and control IgG were not pathogenic. For the samples of all 3 PV patients, depletion of anti-Dsg3/1 IgG resulted in a complete loss of pathogenicity when injected into neonatal mice. In contrast, injection of anti-Dsg3-specific IgG, eluted from the column, induced gross blistering in the mice. Our data clearly show that anti-Dsg3-specific IgG alone is pathogenic in vitro and in vivo, whereas Dsg3/1-depletion results in a complete loss of pathogenicity. Furthermore, our data suggest that Dsg-specific adsorption may be a suitable therapeutic modality to efficiently reduce pathogenic autoantibodies in patients with severe PV.


Asunto(s)
Anticuerpos Antiidiotipos , Autoanticuerpos/inmunología , Desmogleína 3/inmunología , Desmosomas/inmunología , Inmunoglobulina G/inmunología , Pénfigo/inmunología , Adulto , Anciano , Animales , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Antiidiotipos/farmacología , Autoanticuerpos/toxicidad , Desmosomas/patología , Femenino , Humanos , Queratinocitos/inmunología , Queratinocitos/patología , Masculino , Ratones , Pénfigo/patología
8.
J Exp Med ; 215(10): 2554-2566, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30185634

RESUMEN

Cognitive impairment occurs in 40-90% of patients with systemic lupus erythematosus (SLE), which is characterized by autoantibodies to nuclear antigens, especially DNA. We discovered that a subset of anti-DNA antibodies, termed DNRAbs, cross reacts with the N-methyl-d-aspartate receptor (NMDAR) and enhances NMDAR signaling. In patients, DNRAb presence associates with spatial memory impairment. In a mouse model, DNRAb-mediated brain pathology proceeds through an acute phase of excitotoxic neuron loss, followed by persistent alteration in neuronal integrity and spatial memory impairment. The latter pathology becomes evident only after DNRAbs are no longer detectable in the brain. Here we investigate the mechanism of long-term neuronal dysfunction mediated by transient exposure to antibody. We show that activated microglia and C1q are critical mediators of neuronal damage. We further show that centrally acting inhibitors of angiotensin-converting enzyme (ACE) can prevent microglial activation and preserve neuronal function and cognitive performance. Thus, ACE inhibition represents a strong candidate for clinical trials aimed at mitigating cognitive dysfunction.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Anticuerpos Antinucleares/inmunología , Autoanticuerpos , Encéfalo , Lupus Eritematoso Sistémico/inmunología , Trastornos de la Memoria , Neuronas/inmunología , Animales , Autoanticuerpos/inmunología , Autoanticuerpos/toxicidad , Encéfalo/inmunología , Encéfalo/patología , Femenino , Lupus Eritematoso Sistémico/patología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/inmunología , Trastornos de la Memoria/patología , Ratones , Ratones Endogámicos BALB C , Microglía , Neuronas/patología , Receptores de N-Metil-D-Aspartato/inmunología
9.
J Biol Chem ; 292(51): 21180-21192, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29066620

RESUMEN

Tumor cells display on their surface several molecular chaperones that normally reside in the endoplasmic reticulum. Because this display is unique to cancer cells, these chaperones are attractive targets for drug development. Previous epitope-mapping of autoantibodies (AutoAbs) from prostate cancer patients identified the 78-kDa glucose-regulated protein (GRP78) as one such target. Although we previously showed that anti-GRP78 AutoAbs increase tissue factor (TF) procoagulant activity on the surface of tumor cells, the direct effect of TF activation on tumor growth was not examined. In this study, we explore the interplay between the AutoAbs against cell surface-associated GRP78, TF expression/activity, and prostate cancer progression. First, we show that tumor GRP78 expression correlates with disease stage and that anti-GRP78 AutoAb levels parallel prostate-specific antigen concentrations in patient-derived serum samples. Second, we demonstrate that these anti-GRP78 AutoAbs target cell-surface GRP78, activating the unfolded protein response and inducing tumor cell proliferation through a TF-dependent mechanism, a specific effect reversed by neutralization or immunodepletion of the AutoAb pool. Finally, these AutoAbs enhance tumor growth in mice bearing human prostate cancer xenografts, and heparin derivatives specifically abrogate this effect by blocking AutoAb binding to cell-surface GRP78 and decreasing TF expression/activity. Together, these results establish a molecular mechanism in which AutoAbs against cell-surface GRP78 drive TF-mediated tumor progression in an experimental model of prostate cancer. Heparin derivatives counteract this mechanism and, as such, represent potentially appealing compounds to be evaluated in well-designed translational clinical trials.


Asunto(s)
Autoanticuerpos/metabolismo , Membrana Celular/metabolismo , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Tromboplastina/agonistas , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Autoanticuerpos/análisis , Autoanticuerpos/toxicidad , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/inmunología , Membrana Celular/patología , Proliferación Celular/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/uso terapéutico , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Clasificación del Tumor , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/uso terapéutico , Estadificación de Neoplasias , Próstata/efectos de los fármacos , Próstata/inmunología , Próstata/patología , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Distribución Aleatoria , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Propiedades de Superficie , Tromboplastina/análisis , Tromboplastina/metabolismo , Carga Tumoral/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Kidney Int ; 91(5): 1146-1158, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28111009

RESUMEN

Crescentic glomerulonephritis is a life-threatening renal disease that has been extensively studied by the experimental anti-glomerular basement membrane glomerulonephritis (anti-GBM-GN) model. Although T cells have a significant role in this model, athymic/nude mice and rats still develop severe renal disease. Here we further explored the contribution of intrinsic renal cells in the development of T-cell-independent GN lesions. Anti-GBM-GN was induced in three strains of immune-deficient mice (Rag2-/-, Rag2-/-Il2rg-/-, and Rag2-/-Il2rb-/-) that are devoid of either T/B cells or T/B/NK cells. The Rag2-/-Il2rg-/- or Rag2-/-Il2rb-/- mice harbor an additional deletion of either the common gamma chain (γC) or the interleukin-2 receptor ß subunit (IL-2Rß), respectively, impairing IL-15 signaling in particular. As expected, all these strains developed severe anti-GBM-GN. Additionally, bone marrow replenishment experiments allowed us to deduce a protective role for the glomerular-expressed γC during anti-GBM-GN. Given that IL-15 has been found highly expressed in nephritic kidneys despite the absence of lymphocytes, we then studied this cytokine in vitro on primary cultured podocytes from immune-deficient mice (Rag2-/-Il2rg-/- and Rag2-/-Il2rb-/-) compared to controls. IL-15 induced downstream activation of JAK1/3 and SYK in primary cultured podocytes. IL-15-dependent JAK/SYK induction was impaired in the absence of γC or IL-2Rß. We found γC largely induced on podocytes during human glomerulonephritis. Thus, renal lesions are indeed modulated by intrinsic glomerular cells through the γC/IL-2Rß receptor response, to date classically described only in immune cells.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Glomerulonefritis/inmunología , Subunidad gamma Común de Receptores de Interleucina/inmunología , Subunidad beta del Receptor de Interleucina-2/inmunología , Glomérulos Renales/inmunología , Podocitos/inmunología , Animales , Autoanticuerpos/toxicidad , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Glomerulonefritis/inducido químicamente , Glomerulonefritis/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Interleucina-15/inmunología , Interleucina-15/metabolismo , Subunidad beta del Receptor de Interleucina-2/genética , Janus Quinasa 1/metabolismo , Janus Quinasa 3/metabolismo , Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Células Asesinas Naturales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Podocitos/metabolismo , Cultivo Primario de Células , Transducción de Señal , Quinasa Syk/metabolismo
11.
J Mol Neurosci ; 58(4): 470-82, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26746422

RESUMEN

It is well established that the immunization with ocular antigens causes a retinal ganglion cell (RGC) decline, which is accompanied by glia alterations. In this study, the degenerative effects of the immunization with an optic nerve homogenate (ONA) and its purified compound S100 were analyzed on retinas and optic nerves. Since a participation of glia cells in cell death mechanisms is currently discussed, rats were immunized with S100 or ONA. At 14 and 28 days, immune-histological and Western blot analyses were performed to investigate the optic nerve structure (SMI-32), retinal ganglion cells (Brn-3a), apoptosis (cleaved caspase 3, FasL), and glial profile (Iba1, ED1, GFAP, vimentin). Neurofilament dissolution in S100 animals was evident at 14 days (p = 0.047) and increased at 28 days (p = 0.01). ONA optic nerves remained intact at early stages and degenerated later on (p = 0.002). In both groups, RGC loss was detected via immune-histology and Western blot at 28 days (ONA: p = 0.02; S100: p = 0.005). Additionally, more Iba1(+) retinal microglia could be detected at early stages (ONA: p = 0.006; S100: p = 0.028). A slight astrocyte response was detected on Western blots only on ONA retinas (p = 0.01). Hence, the RGC and optic nerve decline was partly antigen dependent, while neuronal loss is paralleled by an early microglial response.


Asunto(s)
Glaucoma/metabolismo , Microglía/metabolismo , Nervio Óptico/patología , Células Ganglionares de la Retina/patología , Animales , Apoptosis , Autoanticuerpos/toxicidad , Autoinmunidad , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Ectodisplasinas/genética , Ectodisplasinas/metabolismo , Glaucoma/etiología , Glaucoma/inmunología , Glaucoma/patología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microglía/patología , Ratas , Ratas Endogámicas Lew , Proteínas S100/inmunología , Vimentina/genética , Vimentina/metabolismo
12.
J Neuroimmunol ; 289: 8-11, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26616866

RESUMEN

Anti-aquaporin-4 autoantibodies are specific for the neuromyelitis optica spectrum disorders (NMOSD) and they have also been described in patients with systemic lupus erythematosus (SLE) with neurological signs consistent with NMOSD. Our objective was to test for the presence and pathogenicity of anti-AQP4 antibodies in SLE patients without neurological disease. Sera from 89 non-CNS-SLE patients were screened for anti-AQP4 autoantibodies. Two of the 89 patients were positive. Archived samples dating back 11 years were also positive. A brain and spinal cord MRI did not reveal any NMOSD-compatible lesions. An in vitro cytotoxicity assay showed that either sera or purified IgG from these patients induced a complement-mediated damage in cultured astrocytes comparable to antibodies obtained from typical NMO patients. We conclude that AQP4-antibodies can be present in SLE patients and persist for many years, without concurrent clinical or radiological NMOSD signs. It is unclear why the anti-AQP4 antibodies did not induce CNS disease.


Asunto(s)
Acuaporina 4/inmunología , Astrocitos/efectos de los fármacos , Autoanticuerpos/sangre , Autoanticuerpos/toxicidad , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/patología , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Lupus Eritematoso Sistémico/inmunología , Masculino , Estadísticas no Paramétricas , Transfección
13.
Int J Mol Sci ; 15(7): 11495-509, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24979132

RESUMEN

Epidemiological studies have demonstrated that women with a history of preeclampsia have a two-fold increased risk of developing cardiovascular diseases in later life. It is not known whether or not this risk is associated with angiotensin II receptor type 1 autoantibody (AT1-AA), an agonist acting via activation of AT1 receptor (AT1R), which is believed to be involved in the pathogenesis of preeclampsia. The objective of the present study was to confirm the hypothesis that AT1-AA exposure during pregnancy may change the maternal cardiac structure and increase the susceptibility of the postpartum heart to ischemia/reperfusion injury (IRI). In the present study, we first established a preeclampsia rat model by intravenous injection of AT1-AA extracted from the plasma of rats immunized with AT1R, observed the susceptibility of the postpartum maternal heart to IRI at 16 weeks postpartum using the Langendorff preparation, and examined the cardiac structure using light and transmission electron microscopy. The modeled animals presented with symptoms very similar to the clinical symptoms of human preeclampsia during pregnancy, including hypertension and proteinuria. The left ventricular weight (LVW) and left ventricular mass index (LVMI) in AT1-AA treatment group were significantly increased as compared with those of the control group (p < 0.01), although there was no significant difference in final weight between the two groups. AT1-AA acting on AT1R not only induced myocardial cell hypertrophy, mitochondrial swelling, cristae disorganization and collagen accumulation in the interstitium but affected the left ventricular (LV) function and delayed recovery from IRI. In contrast, co-treatment with AT1-AA + losartan completely blocked AT1-AA-induced changes in cardiac structure and function. These data indicate that the presence of AT1-AA during pregnancy was strongly associated with the markers of LV geometry changes and remodeling, and increased the cardiac susceptibility to IRI in later life of postpartum maternal rats.


Asunto(s)
Autoanticuerpos/toxicidad , Preeclampsia/inducido químicamente , Receptor de Angiotensina Tipo 1/inmunología , Daño por Reperfusión/etiología , Animales , Colágeno/metabolismo , Femenino , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Losartán/uso terapéutico , Mitocondrias Cardíacas/ultraestructura , Embarazo , Ratas , Ratas Wistar , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/prevención & control , Función Ventricular
14.
Invest Ophthalmol Vis Sci ; 55(7): 4348-59, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24917137

RESUMEN

PURPOSE: Myasthenia gravis demonstrates a distinct predilection for involvement of the extraocular muscles (EOM), and we have hypothesized that this may be due to a unique immunological environment. To assess this hypothesis, we took an unbiased approach to analyze RNA expression profiles in EOM, diaphragm, and extensor digitorum longus (EDL) in rats with experimentally acquired myasthenia gravis (EAMG). METHODS: Experimentally acquired myasthenia gravis was induced in rats by intraperitoneal injection of antibody directed against the acetylcholine receptor (AChR), whereas control rats received antibody known to bind the AChR but not induce disease. After 48 hours, animals were killed and muscles analyzed by RNA expression profiling. Profiling results were validated using qPCR and immunohistochemical analysis. RESULTS: Sixty-two genes common among all muscle groups were increased in expression. These fell into four major categories: 12.8% stress response, 10.5% immune response, 10.5% metabolism, and 9.0% transcription factors. EOM expressed 212 genes at higher levels, not shared by the other two muscles, and a preponderance of EOM gene changes fell into the immune response category. EOM had the most uniquely reduced genes (126) compared with diaphragm (26) and EDL (50). Only 18 downregulated genes were shared by the three muscles. Histological evaluation and disease load index (sum of fold changes for all genes) demonstrated that EOM had the greatest degree of pathology. CONCLUSIONS: Our studies demonstrated that consistent with human myasthenia gravis, EOM demonstrates a distinct RNA expression signature from EDL and diaphragm, which is based on differences in the degree of muscle injury and inflammatory response.


Asunto(s)
Autoinmunidad , Regulación de la Expresión Génica , Músculo Esquelético/metabolismo , Miastenia Gravis Autoinmune Experimental/genética , ARN/genética , Animales , Autoanticuerpos/toxicidad , Femenino , Inmunohistoquímica , Análisis por Micromatrices/métodos , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/metabolismo , Unión Neuromuscular , Ratas , Ratas Endogámicas Lew , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Colinérgicos/inmunología
15.
Transfusion ; 54(12): 3208-15, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24846273

RESUMEN

BACKGROUND: HNA-3a-specific antibodies can cause severe, sometimes fatal, transfusion-related acute lung injury when present in transfused blood. The HNA3-a/b antigens are determined by an R154Q polymorphism in the first of five extracellular (EC) loops of the 10-membrane-spanning choline transporter-like protein 2 (CTL2) expressed on neutrophils, lymphocytes, and other tissues. Approximately 50% of HNA-3a antibodies (Type 1) can be detected using CTL2 Loop 1 peptides containing R154; the remaining 50% (Type 2) fail to recognize this target. Understanding the basis for this difference could guide efforts to develop practical assays to screen blood donors for HNA-3 antibodies. STUDY DESIGN AND METHODS: Reactions of HNA-3a antibodies against recombinant versions of human, mouse, and human/mouse (chimeric) CTL2 were characterized using flow cytometry and various solid-phase assays. RESULTS: The findings show that, for binding to CTL2, Type 2 HNA-3a antibodies require nonpolymorphic amino acid residues in the third, and possibly the second, EC loops of CTL2 to be in a configuration comparable to that found naturally in the cell membrane. In contrast, Type 1 antibodies require only peptides from the first EC loop that contain R154 for recognition. CONCLUSION: Although Type 1 HNA-3a antibodies can readily be detected in solid-phase assays that use a CTL2 peptide containing R154 as a target, development of a practical test to screen blood donors for Type 2 antibodies will pose a serious technical challenge because of the complex nature of the epitope(s) recognized by this antibody subgroup.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Autoanticuerpos/inmunología , Transfusión Sanguínea , Isoantígenos/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas de Transporte de Membrana/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Animales , Autoanticuerpos/toxicidad , Donantes de Sangre , Modelos Animales de Enfermedad , Selección de Donante/métodos , Femenino , Células HEK293 , Humanos , Isoantígenos/genética , Masculino , Glicoproteínas de Membrana/genética , Proteínas de Transporte de Membrana/genética , Ratones , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
16.
PLoS One ; 9(2): e87840, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24505322

RESUMEN

The ß2-adrenergic receptor agonist, albuterol, has been reported beneficial in treating several forms of congenital myasthenia. Here, for the first time, we examined the potential benefit of albuterol in a mouse model of anti-Muscle Specific Kinase (MuSK) myasthenia gravis. Mice received 15 daily injections of IgG from anti-MuSK positive patients, which resulted in whole-body weakness. At neuromuscular junctions in the tibialis anterior and diaphragm muscles the autoantibodies caused loss of postsynaptic acetylcholine receptors, and reduced the amplitudes of the endplate potential and spontaneous miniature endplate potential in the diaphragm muscle. Treatment with albuterol (8 mg/kg/day) during the two-week anti-MuSK injection series reduced the degree of weakness and weight loss, compared to vehicle-treated mice. However, the compound muscle action potential recorded from the gastrocnemius muscle displayed a decremental response in anti-MuSK-injected mice whether treated with albuterol or vehicle. Ongoing albuterol treatment did not increase endplate potential amplitudes compared to vehicle-treated mice nor did it prevent the loss of acetylcholine receptors from motor endplates. On the other hand, albuterol treatment significantly reduced the degree of fragmentation of endplate acetylcholine receptor clusters and increased the extent to which the remaining receptor clusters were covered by synaptophysin-stained nerve terminals. The results provide the first evidence that short-term albuterol treatment can ameliorate weakness in a robust mouse model of anti-MuSK myasthenia gravis. The results also demonstrate that it is possible for albuterol treatment to reduce whole-body weakness without necessarily reversing myasthenic impairment to the structure and function of the neuromuscular junction.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Albuterol/farmacología , Autoanticuerpos/toxicidad , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Animales , Autoanticuerpos/inmunología , Femenino , Humanos , Ratones , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Miastenia Gravis Autoinmune Experimental/inducido químicamente , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/patología , Unión Neuromuscular/inmunología , Unión Neuromuscular/patología , Proteínas Tirosina Quinasas Receptoras/inmunología
17.
J Biol Chem ; 289(7): 4273-85, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24391090

RESUMEN

Development of ectodermal appendages, such as hair, teeth, sweat glands, sebaceous glands, and mammary glands, requires the action of the TNF family ligand ectodysplasin A (EDA). Mutations of the X-linked EDA gene cause reduction or absence of many ectodermal appendages and have been identified as a cause of ectodermal dysplasia in humans, mice, dogs, and cattle. We have generated blocking antibodies, raised in Eda-deficient mice, against the conserved, receptor-binding domain of EDA. These antibodies recognize epitopes overlapping the receptor-binding site and prevent EDA from binding and activating EDAR at close to stoichiometric ratios in in vitro binding and activity assays. The antibodies block EDA1 and EDA2 of both mammalian and avian origin and, in vivo, suppress the ability of recombinant Fc-EDA1 to rescue ectodermal dysplasia in Eda-deficient Tabby mice. Moreover, administration of EDA blocking antibodies to pregnant wild type mice induced in developing wild type fetuses a marked and permanent ectodermal dysplasia. These function-blocking anti-EDA antibodies with wide cross-species reactivity will enable study of the developmental and postdevelopmental roles of EDA in a variety of organisms and open the route to therapeutic intervention in conditions in which EDA may be implicated.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/toxicidad , Anticuerpos Neutralizantes/toxicidad , Autoanticuerpos/toxicidad , Displasia Ectodérmica/inducido químicamente , Displasia Ectodérmica/inmunología , Ectodisplasinas/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales de Origen Murino/genética , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/inmunología , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Secuencia de Bases , Bovinos , Línea Celular , Perros , Displasia Ectodérmica/genética , Displasia Ectodérmica/metabolismo , Displasia Ectodérmica/patología , Ectodisplasinas/genética , Ectodisplasinas/inmunología , Ectodisplasinas/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Embarazo
18.
Circ Res ; 113(1): 78-87, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23788505

RESUMEN

Hypertensive disorders are life-threatening diseases with high morbidity and mortality, affecting billions of individuals worldwide. A multitude of underlying conditions may contribute to hypertension, thus the need for a plethora of treatment options to identify the approach that best meets the needs of individual patients. A growing body of evidence indicates that (1) autoantibodies that bind to and activate the major angiotensin II type I (AT1) receptor exist in the circulation of patients with hypertensive disorders, (2) these autoantibodies contribute to disease pathophysiology, (3) antibody titers correlate to the severity of the disease, and (4) efforts to block or remove these pathogenic autoantibodies have therapeutic potential. These autoantibodies, termed AT1 agonistic autoantibodies have been extensively characterized in preeclampsia, a life-threatening hypertensive condition of pregnancy. As reviewed here, these autoantibodies cause symptoms of preeclampsia when injected into pregnant mice. Somewhat surprisingly, these auto antibodies also appear in 3 animal models of preeclampsia. However, the occurrence of AT1 agonistic autoantibodies is not restricted to pregnancy. These autoantibodies are prevalent among kidney transplant recipients who develop severe transplant rejection and malignant hypertension during the first week after transplantation. AT1 agonistic autoantibodies are also highly abundant among a group of patients with essential hypertension that are refractory to standard therapy. More recently these autoantibodies have been seen in patients with the autoimmune disease, systemic sclerosis. These 3 examples extend the clinical impact of AT1 agonistic autoantibodies beyond pregnancy. Research reviewed here raises the intriguing possibility that preeclampsia and other hypertensive conditions are autoimmune diseases characterized by the presence of pathogenic autoantibodies that activate the major angiotensin receptor, AT1. These pathogenic autoantibodies could serve as presymptomatic biomarkers and therapeutic targets, thereby providing improved medical management for these conditions.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Hipertensión/inmunología , Preeclampsia/inmunología , Receptor de Angiotensina Tipo 1/agonistas , Animales , Antihipertensivos/uso terapéutico , Autoanticuerpos/toxicidad , Biomarcadores , Activación de Complemento , Complemento C3a/inmunología , Citocinas/sangre , Dimerización , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Endotelina-1/sangre , Femenino , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/inmunología , Rechazo de Injerto/inmunología , Humanos , Hipertensión/sangre , Hipertensión/tratamiento farmacológico , Hipertensión/etiología , Hipertensión Maligna/inmunología , Inmunización Pasiva , Trasplante de Riñón/inmunología , Ratones , Placenta/fisiopatología , Complicaciones Posoperatorias/inmunología , Preeclampsia/etiología , Preeclampsia/patología , Embarazo , Receptor de Angiotensina Tipo 1/inmunología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre
19.
J Neuroimmunol ; 261(1-2): 53-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23726906

RESUMEN

The role of antibodies against the P/Q type voltage-gated calcium channels (VGCC-ab) in the pathogenesis of paraneoplastic cerebellar degeneration (PCD) and lung cancer is unclear. We evaluated in mice the effect of intrathecal injection of IgG purified from serum of a patient with both PCD and Lambert-Eaton myasthenic syndrome (LEMS), and from another patient with isolated LEMS. Mice injected with PCD/LEMS IgG developed marked, reversible ataxia compared with those injected with LEMS or control IgG. These findings suggest that P/Q-type VGCC-ab may play a role in the pathogenesis of ataxia in patients with PCD and SCLC.


Asunto(s)
Autoanticuerpos/toxicidad , Canales de Calcio Tipo P/inmunología , Canales de Calcio Tipo Q/inmunología , Ataxia Cerebelosa/inmunología , Degeneración Cerebelosa Paraneoplásica/inmunología , Animales , Canales de Calcio Tipo P/sangre , Ataxia Cerebelosa/inducido químicamente , Células HEK293 , Humanos , Inyecciones Espinales , Ratones , Ratas
20.
Am J Pathol ; 183(2): 402-12, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23747510

RESUMEN

Although it is clear that T helper (Th)17 cells play a pathologic role in the pathogenesis of several inflammatory diseases, the contribution and regulation of pathogenic Th17 cells in the development of glomerulonephritis are still not fully understood. Herein, we show that IL-10-deficient mice exhibit exacerbation of glomerulonephritis after induction with anti-glomerular basement membrane globulin, with enhanced pathogenic Th17 immune responses. We further demonstrate that Rag1(-/-) mice reconstituted with IL-10(-/-) CD4(+) T cells develop more severe glomerulonephritis after induction of anti-glomerular basement membrane disease, with more infiltration of inflammatory cells into the kidneys. Finally, IL-17 and interferon γ double-positive cells were significantly increased in IL-10(-/-) CD4(+) T-cell cultures under pathogenic Th17 conditions compared with wild-type cell cultures. These findings suggest that T-cell-derived IL-10 plays a critical suppressive role in the control of pathogenic Th17 cell differentiation and highlights the importance of IL-10 as protection against glomerulonephritis development.


Asunto(s)
Glomerulonefritis/inmunología , Interleucina-10/deficiencia , Células Th17/patología , Animales , Autoanticuerpos/toxicidad , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Glomerulonefritis/patología , Proteínas de Homeodominio/metabolismo , Inmunidad Celular/inmunología , Interferón gamma/metabolismo , Interleucina-10/fisiología , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Th17/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...