Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 25(4): 671-681, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38448779

RESUMEN

Cognitive impairment is a frequent manifestation of neuropsychiatric systemic lupus erythematosus, present in up to 80% of patients and leading to a diminished quality of life. In the present study, we used a model of lupus-like cognitive impairment that is initiated when antibodies that crossreact with excitatory neuronal receptors penetrate the hippocampus, causing immediate, self-limited, excitotoxic death of hippocampal neurons, which is then followed by a significant loss of dendritic complexity in surviving neurons. This injury creates a maladaptive equilibrium that is sustained in mice for at least 1 year. We identified a feedforward loop of microglial activation and microglia-dependent synapse elimination dependent on neuronal secretion of high mobility group box 1 protein (HMGB1) which binds the receptor for advanced glycation end products (RAGE) and leads to microglial secretion of C1q, upregulation of interleukin-10 with consequent downregulation of leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1), an inhibitory receptor for C1q. Treatment with a centrally acting angiotensin-converting enzyme inhibitor or with an angiotensin-receptor blocker restored a healthy equilibrium, microglial quiescence and intact spatial memory.


Asunto(s)
Autoanticuerpos , Proteína HMGB1 , Animales , Ratones , Complemento C1q , Proteína HMGB1/metabolismo , Enfermedades Neuroinflamatorias , Calidad de Vida , Receptor para Productos Finales de Glicación Avanzada/metabolismo
2.
Res Sq ; 2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37292843

RESUMEN

Cognitive impairment is a frequent manifestation of neuropsychiatric systemic lupus erythematosus (NPSLE), present in up to 80% of patients and leading to a diminished quality of life. We have developed a model of lupus-like cognitive impairment which is initiated when anti-DNA, anti-N-methyl D-aspartate receptor (NMDAR) cross- reactive antibodies, which are present in 30% of SLE patients, penetrate the hippocampus1. This leads to immediate, self-limited excitotoxic death of CA1 pyramidal neurons followed by a significant loss of dendritic arborization in the remaining CA1 neurons and impaired spatial memory. Both microglia and C1q are required for dendritic loss1. Here we show that this pattern of hippocampal injury creates a maladaptive equilibrium that is sustained for at least one year. It requires HMGB1 secretion by neurons to bind RAGE, a receptor for HMGB1 expressed on microglia, and leads to decreased expression of microglial LAIR-1, an inhibitory receptor for C1q. The angiotensin converting enzyme (ACE) inhibitor captopril, which can restore a healthy equilibrium, microglial quiescence, and intact spatial memory, leads to upregulation of LAIR-1. This paradigm highlights HMGB1:RAGE and C1q:LAIR-1 interactions as pivotal pathways in the microglial-neuronal interplay that defines a physiologic versus a maladaptive equilibrium.

4.
Nat Commun ; 11(1): 1403, 2020 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-32179753

RESUMEN

Patients with Systemic lupus erythematosus (SLE) experience various peripheral and central nervous system manifestations including spatial memory impairment. A subset of autoantibodies (DNRAbs) cross-react with the GluN2A and GluN2B subunits of the NMDA receptor (NMDAR). We find that these DNRAbs act as positive allosteric modulators on NMDARs with GluN2A-containing NMDARs, even those containing a single GluN2A subunit, exhibiting a much greater sensitivity to DNRAbs than those with exclusively GluN2B. Accordingly, GluN2A-specific antagonists provide greater protection from DNRAb-mediated neuronal cell death than GluN2B antagonists. Using transgenic mice to perturb expression of either GluN2A or GluN2B in vivo, we find that DNRAb-mediated disruption of spatial memory characterized by early neuronal cell death and subsequent microglia-dependent pathologies requires GluN2A-containing NMDARs. Our results indicate that GluN2A-specific antagonists or negative allosteric modulators are strong candidates to treat SLE patients with nervous system dysfunction.


Asunto(s)
Autoanticuerpos/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/psicología , Receptores de N-Metil-D-Aspartato/inmunología , Memoria Espacial , Animales , Muerte Celular , Femenino , Humanos , Lupus Eritematoso Sistémico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/inmunología , Receptores de N-Metil-D-Aspartato/genética
5.
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
6.
Front Immunol ; 9: 2824, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619247

RESUMEN

A primary mechanism for activation of innate immunity is recognition of damage or pathogen associated molecular patterns by pattern recognition receptors (PRRs). Nucleic acid is a damage associated molecular pattern molecule that when internalized into a monocyte and recognized by intracellular nucleic acid sensing toll like receptors will cause production of type 1 interferon. The process by which DNA or RNA is delivered into the cytosol of monocytes in systemic lupus erythematosus remains incompletely understood, and therapeutic approaches to prevent DNA-mediated monocyte activation are needed. We identified two mechanisms for internalization of DNA by monocytes. IgG-bound DNA was internalized by interacting with Fc gamma receptor IIa, while high-mobility group box-1 protein-bound DNA was internalized by interacting with the receptor for advanced glycation end products. Both pathways contribute to an inflammatory phenotype in monocytes exposed to serum from patients with SLE. Moreover, both of these pathways can be inhibited by a pentapeptide, DWEYS, which is a DNA mimetope. In one instance DWEYS directly competes with DNA for antibody binding and in the other DWEYS binds high-mobility group box-1 and blocks its interaction with RAGE. Our data highlight distinct pathways involved in nucleic acid enters monocytes in SLE, and identify a potential therapeutic to prevent nucleic acid internalization in SLE.


Asunto(s)
Anticuerpos Antinucleares , Ácidos Nucleicos Libres de Células , Inmunoglobulina G , Interferones , Lupus Eritematoso Sistémico , Monocitos , Animales , Anticuerpos Antinucleares/sangre , Anticuerpos Antinucleares/inmunología , Ácidos Nucleicos Libres de Células/sangre , Ácidos Nucleicos Libres de Células/inmunología , Femenino , Proteína HMGB1/sangre , Proteína HMGB1/inmunología , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Interferones/sangre , Interferones/inmunología , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Masculino , Ratones Mutantes , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , omegacloroacetofenona
7.
Exp Ther Med ; 14(1): 228-238, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28672919

RESUMEN

The potential role of systemic inflammation on brain injury in newborns with congenital heart disease (CHD) was assessed by measuring levels of central nervous system (CNS)-derived proteins in serum prior to and following cardiac surgery. A total of 23 newborns (gestational age, 39±1 weeks) with a diagnosis of CHD that required cardiac surgery with cardiopulmonary bypass (CPB) were enrolled in the current study. Serum samples were collected immediately prior to surgery and 2, 24 and 48 h following CPB, and serum levels of phosphorylated neurofilament-heavy subunit (pNF-H), neuron-specific enolase (NSE) and S100B were analyzed. Systemic inflammation was assessed by measuring serum concentrations of complement C5a and complement sC5b9, and the following cytokines: Interleukin (IL)-1ß, IL-6, IL-8, IL-10, IL12p70, interferon γ and tumor necrosis factor (TNF)-α. Analysis of cord blood from normal term deliveries (n=26) provided surrogate normative values for newborns. pNF-H and S100B were 2.4- to 2.8-fold higher (P<0.0001) in patient sera than in cord blood prior to surgery and remained elevated following CPB. Pre-surgical serum pNF-H and S100B levels directly correlated with interleukin (IL)-12p70 (ρ=0.442, P<0.05). pNF-H was inversely correlated with arterial pO2 prior to surgery (ρ=-0.493, P=0.01) and directly correlated with arterial pCO2 post-CPB (ρ=0.426, P<0.05), suggesting that tissue hypoxia and inflammation contribute to blood brain barrier (BBB) dysfunction and neuronal injury. Serum IL12p70, IL-6, IL-8, IL-10 and TNF-α levels were significantly higher in patients than in normal cord blood and levels of these cytokines increased following CPB (P<0.001). Activation of complement was observed in all patients prior to surgery, and serum C5a and sC5b9 remained elevated up to 48 h post-surgery. Furthermore, they were correlated (P<0.05) with low arterial pO2, high pCO2 and elevated arterial pressure in the postoperative period. Length of mechanical ventilation was associated directly with post-surgery serum IL-12p70 and IL-8 concentrations (P<0.05). Elevated serum concentrations of pNF-H and S100B in neonates with CHD suggest BBB dysfunction and CNS injury, with concurrent hypoxemia and an activated inflammatory response potentiating this effect.

8.
Discov Med ; 22(121): 201-207, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27875671

RESUMEN

Cells and molecules of the immune system contribute to brain pathology as well as to brain homeostasis. We suggest that there are numerous anti-brain antibodies that can cause acute neuronal dysfunction if they penetrate brain parenchyma. Many of these acute immune-mediated insults may alter the homeostatic mechanisms in the brain and initiate pathologic events that no longer depend on the presence of the inciting antibody, but rather on microglial cell activation. This paradigm, if correct, suggests that there may be two potential moments of therapeutic intervention. The first moment is when antibody contacts cells of the central nervous system and the second is when microglia become activated and impair normal neuronal functions. In this review, we discuss data that support this model for immune-mediated pathology in both the adult brain and the developing fetal brain.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Encefalopatías/inmunología , Encéfalo/inmunología , Microglía/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/patología , Encéfalo/patología , Encefalopatías/patología , Humanos , Microglía/patología
9.
J Med Chem ; 59(19): 8859-8867, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27603688

RESUMEN

Systemic lupus erythematosus is an autoimmune disease that can affect numerous tissues and is characterized by the production of nuclear antigen-directed autoantibodies (e.g., anti-dsDNA). Using a combination of virtual and ELISA-based screens, we made the intriguing discovery that several HIV-protease inhibitors can function as decoy antigens to specifically inhibit the binding of anti-dsDNA antibodies to target antigens such as dsDNA and pentapeptide DWEYS. Computational modeling revealed that HIV-protease inhibitors comprised structural features present in DWEYS and predicted that analogues containing more flexible backbones would possess preferred binding characteristics. To address this, we reduced the internal amide backbone to improve flexibility, producing new small-molecule decoy antigens, which neutralize anti-dsDNA antibodies in vitro, in situ, and in vivo. Pharmacokinetic and SLE model studies demonstrated that peptidomimetic FISLE-412,1 a reduced HIV protease inhibitor analogue, was well-tolerated, altered serum reactivity to DWEYS, reduced glomeruli IgG deposition, preserved kidney histology, and delayed SLE onset in NZB/W F1 mice.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Animales , ADN/inmunología , Descubrimiento de Drogas , Femenino , Inhibidores de la Proteasa del VIH/farmacocinética , Inhibidores de la Proteasa del VIH/farmacología , Humanos , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos NZB , Modelos Moleculares
10.
Immunol Res ; 63(1-3): 18-25, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26507553

RESUMEN

We briefly review the protective role of maternal antibodies during fetal development and at early postnatal stages. We describe antibody delivery to fetuses, particularly in the context of the developing blood-brain barrier (BBB), and present the essential concepts regarding the adult BBB, together with existing information on the prenatal developing BBB. We focus on maternal antibody transfer to the developing brain and the consequences of the presence of pathogenic antibodies at early stages of brain development on subsequent brain dysfunction.


Asunto(s)
Anticuerpos/metabolismo , Barrera Hematoencefálica/inmunología , Encefalopatías/inmunología , Encéfalo/inmunología , Inmunidad Materno-Adquirida , Adulto , Animales , Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Encéfalo/embriología , Femenino , Humanos , Exposición Materna/efectos adversos , Embarazo
11.
Trends Immunol ; 36(11): 709-724, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26494046

RESUMEN

The brain is normally sequestered from antibody exposure by the blood brain barrier. However, antibodies can access the brain during fetal development before the barrier achieves full integrity, and in disease states when barrier integrity is compromised. Recent studies suggest that antibodies contribute to brain pathology associated with autoimmune diseases such as systemic lupus erythematosus and neuromyelitis optica, and can lead to transient or permanent behavioral or cognitive abnormalities. We review these findings here and examine the circumstances associated with antibody entry into the brain, the routes of access and the mechanisms that then effect pathology. Understanding these processes and the nature and specificity of neuronal autoantibodies may reveal therapeutic strategies toward alleviating or preventing the neurological pathologies and behavioral abnormalities associated with autoimmune disease.


Asunto(s)
Anticuerpos/inmunología , Encéfalo/inmunología , Encéfalo/patología , Animales , Humanos
12.
EBioMedicine ; 2(7): 755-64, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26286205

RESUMEN

Patients with systemic lupus erythematosus (SLE) experience cognitive abnormalities in multiple domains including processing speed, executive function, and memory. Here we show that SLE patients carrying antibodies that bind DNA and the GluN2A and GluN2B subunits of the N-methyl-d-aspartate receptor (NMDAR), termed DNRAbs, displayed a selective impairment in spatial recall. Neural recordings in a mouse model of SLE, in which circulating DNRAbs penetrate the hippocampus, revealed that CA1 place cells exhibited a significant expansion in place field size. Structural analysis showed that hippocampal pyramidal cells had substantial reductions in their dendritic processes and spines. Strikingly, these abnormalities became evident at a time when DNRAbs were no longer detectable in the hippocampus. These results suggest that antibody-mediated neurocognitive impairments may be highly specific, and that spatial cognition may be particularly vulnerable to DNRAb-mediated structural and functional injury to hippocampal cells that evolves after the triggering insult is no longer present.


Asunto(s)
Autoanticuerpos/metabolismo , Cognición , Receptores de N-Metil-D-Aspartato/inmunología , Percepción Espacial , Adulto , Animales , Anticuerpos Antinucleares/inmunología , Membrana Celular/metabolismo , Dendritas/metabolismo , Femenino , Células HEK293 , Hipocampo/patología , Humanos , Lupus Eritematoso Sistémico/inmunología , Masculino , Ratones Endogámicos BALB C , Persona de Mediana Edad , Células Piramidales/metabolismo , Memoria Espacial
13.
Sci Transl Med ; 6(263): 263ra158, 2014 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-25411471

RESUMEN

Pivotal to brain development and function is an intact blood-brain barrier (BBB), which acts as a gatekeeper to control the passage and exchange of molecules and nutrients between the circulatory system and the brain parenchyma. The BBB also ensures homeostasis of the central nervous system (CNS). We report that germ-free mice, beginning with intrauterine life, displayed increased BBB permeability compared to pathogen-free mice with a normal gut flora. The increased BBB permeability was maintained in germ-free mice after birth and during adulthood and was associated with reduced expression of the tight junction proteins occludin and claudin-5, which are known to regulate barrier function in endothelial tissues. Exposure of germ-free adult mice to a pathogen-free gut microbiota decreased BBB permeability and up-regulated the expression of tight junction proteins. Our results suggest that gut microbiota-BBB communication is initiated during gestation and propagated throughout life.


Asunto(s)
Barrera Hematoencefálica , Intestinos/microbiología , Microbiota , Animales , Femenino , Ratones , Permeabilidad , Embarazo , Uniones Estrechas/metabolismo
14.
J Cereb Blood Flow Metab ; 34(8): 1315-20, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24824914

RESUMEN

Systemic lupus erythematosus (SLE) is characterized by multiorgan inflammation, neuropsychiatric disorders (NPSLE), and anti-nuclear antibodies. We previously identified a subset of anti-DNA antibodies (DNRAb) cross-reactive with the N-methyl-D-aspartate receptor, present in 30% to 40% of patients, able to enhance excitatory post-synaptic potentials and trigger neuronal apoptosis. DNRAb+ mice exhibit memory impairment or altered fear response, depending on whether the antibody penetrates the hippocampus or amygdala. Here, we used 18F-fluorodeoxyglucose (FDG) microPET to plot changes in brain metabolism after regional blood-brain barrier (BBB) breach. In DNRAb+ mice, metabolism declined at the site of BBB breach in the first 2 weeks and increased over the next 2 weeks. In contrast, DNRAb- mice exhibited metabolic increases in these regions over the 4 weeks after the insult. Memory impairment was present in DNRAb+ animals with hippocampal BBB breach and altered fear conditioning in DNRAb+ mice with amygdala BBB breach. In DNRAb+ mice, we observed an inverse relationship between neuron number and regional metabolism, while a positive correlation was observed in DNRAb- mice. These findings suggest that local metabolic alterations in this model take place through different mechanisms with distinct time courses, with important implications for the interpretation of imaging data in SLE subjects.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Anticuerpos Antinucleares/metabolismo , Barrera Hematoencefálica/metabolismo , Hipocampo/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Amígdala del Cerebelo/diagnóstico por imagen , Amígdala del Cerebelo/patología , Animales , Conducta Animal , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Femenino , Fluorodesoxiglucosa F18 , Hipocampo/diagnóstico por imagen , Hipocampo/patología , Lupus Eritematoso Sistémico/diagnóstico por imagen , Lupus Eritematoso Sistémico/patología , Lupus Eritematoso Sistémico/psicología , Ratones , Ratones Endogámicos BALB C , Neuronas/metabolismo , Neuronas/patología , Tomografía de Emisión de Positrones
15.
Methods Mol Biol ; 900: 181-206, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22933070

RESUMEN

This chapter describes methods utilized in establishing a mouse model of neuropsychiatric lupus encompassing both cognitive and emotional dysfunction, and a model of the influence of maternal antibody on the developing brain. The antibody of interest binds the N-methyl-D: -aspartate receptor (NMDAR), a receptor for glutamate that is a major excitatory neurotransmitter in the brain involved in synaptic plasticity, in memory and learning, and in emotional responses.We introduce basic concepts of these models and provide protocols for the following: (1) the induction of anti-dsDNA, anti-NMDAR antibodies, (2) testing serum antibody titer by ELISA, (3) breaching blood brain barrier (BBB) integrity with LPS and epinephrine, (4) passive transfer of pathology by injecting human and mouse brain-reactive antibodies into adult mouse as well as injecting the antibody into gestating mice and transfer of antibody from dam to fetus, (5) blocking NMDAR-mediated pathogenicity in vivo, (6) evacuation of blood from the brain by cardiac perfusion to preserve the brain for histology, (7) evaluating injured/apoptotic neurons in brain histology, (8) preparing membrane-enriched brain -fractions for NMDAR analysis.


Asunto(s)
Autoanticuerpos/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Lupus Eritematoso Sistémico/inmunología , Biología Molecular/métodos , Receptores de N-Metil-D-Aspartato/inmunología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Epinefrina/farmacología , Femenino , Humanos , Inmunización , Inmunización Pasiva , Lipopolisacáridos/farmacología , Lupus Eritematoso Sistémico/patología , Membranas/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Miocardio/patología , Perfusión
16.
J Exp Med ; 209(6): 1083-9, 2012 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-22565825

RESUMEN

Systemic lupus erythematosus (SLE), a disease of women during childbearing years, is characterized by the production of double-stranded DNA antibodies. A subset of these antibodies, present in 40% of patients, cross-reacts with the NR2A and NR2B subunits of the N-methyl-d-aspartate receptor (NMDAR). In this study, we show that, in mouse models, these antibodies cause a loss of female fetus viability by inducing apoptosis of NR2A-expressing neurons within the brainstem late in fetal development; gender specificity derives from a time-dependent increased expression of NR2A in female brainstem or increased vulnerability of female fetal neurons to signaling through NR2A-containing NMDARs. This paradigm is consistent with available data on the sex ratio of live births of women with SLE. It represents a novel mechanism by which maternal autoantibodies can severely affect fetal health in a gender-specific fashion and raises the question of how many maternal antibodies affect brain development or exhibit gender-specific fetal effects.


Asunto(s)
Autoanticuerpos/inmunología , Encéfalo/embriología , Muerte Fetal/inmunología , Animales , Animales Recién Nacidos , Anticuerpos Antinucleares/inmunología , Apoptosis/genética , Apoptosis/inmunología , Encéfalo/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Lupus Eritematoso Sistémico/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Neuronas/inmunología , Oligopéptidos/inmunología , Oligopéptidos/farmacología , Embarazo , Receptores de N-Metil-D-Aspartato/genética , Factores Sexuales , Razón de Masculinidad
17.
Proc Natl Acad Sci U S A ; 107(43): 18569-74, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20921396

RESUMEN

Damaging interactions between antibodies and brain antigenic targets may be responsible for an expanding range of neurological disorders. In the case of systemic lupus erythematosus (SLE), patients generate autoantibodies (AAbs) that frequently bind dsDNA. Although some symptoms of SLE may arise from direct reactivity to dsDNA, much of the AAb-mediated damage originates from cross-reactivity with other antigens. We have studied lupus AAbs that bind dsDNA and cross-react with the NR2A and NR2B subunits of the NMDA receptor (NMDAR). In adult mouse models, when the blood-brain barrier is compromised, these NMDAR-reactive AAbs access the brain and elicit neuronal death with ensuing cognitive dysfunction and emotional disturbance. The cellular mechanisms that underlie these deleterious effects remain incompletely understood. Here, we show that, at low concentration, the NMDAR-reactive AAbs are positive modulators of receptor function that increase the size of NMDAR-mediated excitatory postsynaptic potentials, whereas at high concentration, the AAbs promote excitotoxicity through enhanced mitochondrial permeability transition. Other synaptic receptors are completely unaffected by the AAbs. NMDAR activation is required for producing both the synaptic and the mitochondrial effects. Our study thus reveals the mechanisms by which NMDAR-reactive AAbs trigger graded cellular alterations, which are likely to be responsible for the transient and permanent neuropsychiatric symptoms observed in patients with SLE. Our study also provides a model in which local AAb concentration determines the exact nature of the cellular response.


Asunto(s)
Autoanticuerpos/toxicidad , Encéfalo/inmunología , Encéfalo/fisiopatología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/fisiopatología , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Vasculitis por Lupus del Sistema Nervioso Central/fisiopatología , Neurotoxinas/toxicidad , Animales , Reacciones Cruzadas , Potenciales Postsinápticos Excitadores , Femenino , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Poro de Transición de la Permeabilidad Mitocondrial , Modelos Inmunológicos , Modelos Neurológicos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/inmunología
18.
J Exp Med ; 206(10): 2151-9, 2009 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-19752183

RESUMEN

The unfolded protein response (UPR) is a stress response pathway that is driven by the increased load of unfolded proteins in the endoplasmic reticulum of highly secretory cells such as plasma cells (PCs). X box binding protein 1 (XBP1) is a transcription factor that mediates one branch of the UPR and is crucial for the development of antibody-secreting PCs. PCs represent only one class of terminally differentiated B cells, however, and little is known about the role for XBP1 in the other class: memory B cells. We have developed an XBP1(fl/fl) CD19(+/cre) conditional knockout (XBP1(CD19)) mouse to build upon our current understanding of the function of XBP1 in PC differentiation as well as to explore the role of XBP1 in memory cell development. Using this model, we show that XBP1(CD19) mice are protected from disease in an autoantibody-mediated mouse lupus model. We also identify a novel developmental stage at which B cells express the traditional PC marker CD138 (syndecan-1) but have yet to undergo XBP1-dependent functional and morphological differentiation into antibody-secreting cells. Finally, we show that memory B cells develop normally in XBP1(CD19) mice, demonstrating that XBP1-mediated functions occur independently of any memory cell lineage commitment.


Asunto(s)
Linfocitos B/fisiología , Proteínas de Unión al ADN/fisiología , Células Plasmáticas/citología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Memoria Inmunológica , Lupus Eritematoso Sistémico/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos MRL lpr , Ratones Endogámicos NZB , Ratones Noqueados , Factores de Transcripción del Factor Regulador X , Sindecano-1/análisis , Proteína 1 de Unión a la X-Box
19.
Nat Rev Immunol ; 9(6): 449-56, 2009 06.
Artículo en Inglés | MEDLINE | ID: mdl-19424277

RESUMEN

We propose that the normal immunocompetent B cell repertoire is replete with B cells making antibodies that recognize brain antigens. Although B cells that are reactive with self antigen are normally silenced during B cell maturation, the blood-brain barrier (BBB) prevents many brain antigens from participating in this process. This enables the generation of a B cell repertoire that is sufficiently diverse to cope with numerous environmental challenges. It requires, however, that the integrity of the BBBs is uninterrupted throughout life to protect the brain from antibodies that crossreact with microorganisms and brain antigens. Under conditions of BBB compromise, and during fetal development, we think that these antibodies can alter brain function in otherwise healthy individuals.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Barrera Hematoencefálica/inmunología , Encéfalo/fisiología , Animales , Encéfalo/inmunología , Humanos , Receptores de N-Metil-D-Aspartato/inmunología , Receptores de N-Metil-D-Aspartato/metabolismo
20.
Nat Med ; 15(1): 91-6, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19079257

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease mediated by autoantibodies and preferentially affecting women of childbearing age. Because the offspring of mothers with SLE show a high frequency of learning disorders, we hypothesized that maternally transferred autoantibodies that bind DNA and the N-methyl-D-aspartate receptor (NMDAR) could have a pathogenic role during fetal brain development. Here we describe a maternal SLE mouse model wherein pregnant dams harbored DNA-specific, NMDAR-specific autoantibodies throughout gestation. High titers of these autoantibodies in maternal circulation led to histological abnormalities in fetal brain and subsequent cognitive impairments in adult offspring. These data support a paradigm in which in utero exposure to neurotoxic autoantibodies causes abnormal brain development with long-term consequences. This paradigm may apply to multiple congenital neuropsychiatric disorders.


Asunto(s)
Autoanticuerpos/inmunología , Lupus Eritematoso Sistémico/inmunología , Malformaciones del Sistema Nervioso/etiología , Neuronas/inmunología , Complicaciones del Embarazo/inmunología , Animales , Anticuerpos Antinucleares/inmunología , Autoanticuerpos/sangre , Citotoxinas/inmunología , Modelos Animales de Enfermedad , Femenino , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/complicaciones , Intercambio Materno-Fetal/inmunología , Ratones , Ratones Endogámicos BALB C , Malformaciones del Sistema Nervioso/inmunología , Neuronas/metabolismo , Embarazo , Complicaciones del Embarazo/sangre , Receptores de N-Metil-D-Aspartato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...