Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 213(2): 214-225, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38829123

RESUMEN

The interactions between chemokines and their receptors, particularly in the context of inflammation, are complex, with individual receptors binding multiple ligands and individual ligands interacting with multiple receptors. In addition, there are numerous reports of simultaneous coexpression of multiple inflammatory chemokine receptors on individual inflammatory leukocyte subtypes. Overall, this has previously been interpreted as redundancy and proposed as a protective mechanism to ensure that the inflammatory response is robust. By contrast, we have hypothesized that the system is not redundant but exquisitely subtle. Our interests relate to the receptors CCR1, CCR2, CCR3, and CCR5, which, together, regulate nonneutrophilic myeloid cell recruitment to inflammatory sites. In this study, we demonstrate that although most murine monocytes exclusively express CCR2, there is a small subpopulation that is expanded during inflammation and coexpresses CCR1 and CCR2. Combinations of transcript and functional analysis demonstrate that this is not redundant expression and that coexpression of CCR1 and CCR2 marks a phenotypically distinct population of monocytes characterized by expression of genes otherwise typically associated with neutrophils. Single-cell RNA sequencing confirms this as a monodisperse population of atypical monocytes. This monocytic population has previously been described as having immunosuppressive activity. Overall, our data confirm combinatorial chemokine receptor expression by a subpopulation of monocytes but demonstrate that this is not redundant expression and marks a discrete monocytic population.


Asunto(s)
Monocitos , Receptores CCR1 , Receptores CCR2 , Receptores CCR1/genética , Receptores CCR1/metabolismo , Receptores CCR2/genética , Receptores CCR2/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Inflamación/inmunología
2.
J Peripher Nerv Syst ; 28(1): 17-31, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36710500

RESUMEN

The major determinant of poor outcome in Guillain-Barré syndrome (GBS) is axonal degeneration. Pathways leading to primary axonal injury in the motor axonal variant are well established, whereas mechanisms of secondary axonal injury in acute inflammatory demyelinating polyneuropathy (AIDP) are unknown. We recently developed an autoantibody-and complement-mediated model of murine AIDP, in which prominent injury to glial membranes at the node of Ranvier results in severe disruption to paranodal components. Acutely, axonal integrity was maintained, but over time secondary axonal degeneration occurred. Herein, we describe the differential mechanisms underlying acute glial membrane injury and secondary axonal injury in this model. Ex vivo nerve-muscle explants were injured for either acute or extended periods with an autoantibody-and complement-mediated injury to glial paranodal membranes. This model was used to test several possible mechanisms of axon degeneration including calpain activation, and to monitor live axonal calcium signalling. Glial calpains induced acute disruption of paranodal membrane proteins in the absence of discernible axonal injury. Over time, we observed progressive axonal degeneration which was markedly attenuated by axon-specific calpain inhibition. Injury was unaffected by all other tested methods of protection. Trans-axolemmal diffusion of fluorescent proteins  and live calcium imaging studies indirectly demonstrated the presence of nanoruptures in the axon membrane. This study outlines one mechanism by which secondary axonal degeneration arises in the AIDP variant of GBS where acute paranodal loop injury is prominent. The data also support the development of calpain inhibitors to attenuate both primary and secondary axonal degeneration in GBS.


Asunto(s)
Síndrome de Guillain-Barré , Humanos , Ratones , Animales , Calcio , Calpaína , Axones , Autoanticuerpos
3.
J Peripher Nerv Syst ; 28(1): 4-16, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36335586

RESUMEN

Axon degeneration accounts for the poor clinical outcome in Guillain-Barré syndrome (GBS), yet no treatments target this key pathogenic stage. Animal models demonstrate anti-ganglioside antibodies (AGAb) induce axolemmal complement pore formation through which calcium flux activates the intra-axonal calcium-dependent proteases, calpains. We previously showed protection of axonal components using soluble calpain inhibitors in ex vivo GBS mouse models, and herein, we assess the potential of axonally-restricted calpain inhibition as a neuroprotective therapy operating in vivo. Using transgenic mice that over-express the endogenous human calpain inhibitor calpastatin (hCAST) neuronally, we assessed distal motor nerve integrity in our established GBS models. We induced immune-mediated injury with monoclonal AGAb plus a source of human complement. The calpain substrates neurofilament and AnkyrinG, nerve structural proteins, were assessed by immunolabelling and in the case of neurofilament, by single-molecule arrays (Simoa). As the distal intramuscular portion of the phrenic nerve is prominently targeted in our in vivo model, respiratory function was assessed by whole-body plethysmography as the functional output in the acute and extended models. hCAST expression protects distal nerve structural integrity both ex and in vivo, as shown by attenuation of neurofilament breakdown by immunolabelling and Simoa. In an extended in vivo model, while mice still initially undergo respiratory distress owing to acute conduction failure, the recovery phase was accelerated by hCAST expression. Axonal calpain inhibition can protect the axonal integrity of the nerve in an in vivo GBS paradigm and hasten recovery. These studies reinforce the strong justification for developing further animal and human clinical studies using exogenous calpain inhibitors.


Asunto(s)
Síndrome de Guillain-Barré , Ratones , Humanos , Animales , Calpaína/metabolismo , Calcio/metabolismo , Axones/patología , Ratones Transgénicos
4.
Clin Auton Res ; 33(6): 767-775, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37943335

RESUMEN

PURPOSE: Acute decompensated heart failure (ADHF) is associated with inflammation, oxidative stress, and excess sympathetic drive. It is unknown whether neuromodulation would improve inflammation and oxidative stress in acute heart failure. We, therefore, performed this proof-of-concept study to evaluate the effects of neuromodulation using noninvasive low-level tragus stimulation on inflammation and oxidative stress in ADHF. METHODS: Nineteen patients with ejection fraction < 40% were randomized to neuromodulation 4 h twice daily (6-10 a.m. and 6-10 p.m.) (n = 8) or sham stimulation (n = 11) during hospital admission. All patients received standard-of-care treatment. Blood samples were collected at admission and discharge. Serum cytokines were assayed using standard immunosorbent techniques. Reactive oxygen species inducibility from cultured coronary endothelial cells exposed to patient sera was determined using a dihydrodichlorofluorescein probe test (expressed as fluorescein units). RESULTS: Compared to sham stimulation, neuromodulation was associated with a significant reduction of circulating serum interleukin-6 levels (-78% vs. -9%; p = 0.012). Similarly, neuromodulation led to a reduction of endothelial cell oxidative stress in the neuromodulation group (1363 units to 978 units, p = 0.003) compared to sham stimulation (1146 units to 1083 units, p = 0.094). No significant differences in heart rate, blood pressure, or renal function were noted between the two groups. CONCLUSION: In this proof-of-concept pilot study, in acute decompensated heart failure, neuromodulation was feasible and safe and was associated with a reduction in systemic inflammation and attenuation of coronary endothelial cellular oxidative stress. CLINICAL TRIAL REGISTRATION: NCT02898181.


Asunto(s)
Células Endoteliales , Insuficiencia Cardíaca , Humanos , Proyectos Piloto , Insuficiencia Cardíaca/terapia , Inflamación/terapia , Estrés Oxidativo
5.
J Neurosci ; 39(1): 63-77, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30446529

RESUMEN

Sulfatides and gangliosides are raft-associated glycolipids essential for maintaining myelinated nerve integrity. Mice deficient in sulfatide (cerebroside sulfotransferase knock-out, CST-/-) or complex gangliosides (ß-1,4-N-acetylegalactosaminyltransferase1 knock-out, GalNAc-T-/-) display prominent disorganization of proteins at the node of Ranvier (NoR) in early life and age-dependent neurodegeneration. Loss of neuronal rather than glial complex gangliosides underpins the GalNAc-T-/- phenotype, as shown by neuron- or glial-specific rescue, whereas sulfatide is principally expressed and functional in glial membranes. The similarities in NoR phenotype of CST-/-, GalNAc-T-/-, and axo-glial protein-deficient mice suggests that these glycolipids stabilize membrane proteins including neurofascin155 (NF155) and myelin-associated glycoprotein (MAG) at axo-glial junctions. To assess the functional interactions between sulfatide and gangliosides, CST-/- and GalNAc-T-/- genotypes were interbred. CST-/-× GalNAc-T-/- mice develop normally to postnatal day 10 (P10), but all die between P20 and P25, coinciding with peak myelination. Ultrastructural, immunohistological, and biochemical analysis of either sex revealed widespread axonal degeneration and disruption to the axo-glial junction at the NoR. In addition to sulfatide-dependent loss of NF155, CST-/- × GalNAc-T-/- mice exhibited a major reduction in MAG protein levels in CNS myelin compared with WT and single-lipid-deficient mice. The CST-/- × GalNAc-T-/- phenotype was fully restored to that of CST-/- mice by neuron-specific expression of complex gangliosides, but not by their glial-specific expression nor by the global expression of a-series gangliosides. These data indicate that sulfatide and complex b-series gangliosides on the glial and neuronal membranes, respectively, act in concert to promote NF155 and MAG in maintaining the stable axo-glial interactions essential for normal nerve function.SIGNIFICANCE STATEMENT Sulfatides and complex gangliosides are membrane glycolipids with important roles in maintaining nervous system integrity. Node of Ranvier maintenance in particular requires stable compartmentalization of multiple membrane proteins. The axo-glial adhesion molecules neurofascin155 (NF155) and myelin-associated glycoprotein (MAG) require membrane microdomains containing either sulfatides or complex gangliosides to localize and function effectively. The cooperative roles of these microdomains and associated proteins are unknown. Here, we show vital interdependent roles for sulfatides and complex gangliosides because double (but not single) deficiency causes a rapidly lethal phenotype at an early age. These findings suggest that sulfatides and complex gangliosides on opposing axo-glial membranes are responsible for essential tethering of the axo-glial junction proteins NF155 and MAG, which interact to maintain the nodal complex.


Asunto(s)
Axones/fisiología , Gangliósidos/metabolismo , Gangliósidos/fisiología , Vaina de Mielina/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Sulfoglicoesfingolípidos/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Femenino , Genotipo , Esperanza de Vida , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/fisiología , N-Acetilgalactosaminiltransferasas/genética , Factores de Crecimiento Nervioso/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Nódulos de Ranvier/fisiología , Sulfotransferasas/genética , Sulfotransferasas/fisiología
6.
J Peripher Nerv Syst ; 25(2): 143-151, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32250537

RESUMEN

In mouse models of acute motor axonal neuropathy, anti-ganglioside antibodies (AGAbs) bind to motor axons, notably the distal nerve, and activate the complement cascade. While complement activation is well studied in this model, the role of inflammatory cells is unknown. Herein we aimed to investigate the contribution of phagocytic cells including macrophages, neutrophils and perisynaptic Schwann cells (pSCs) to distal nerve pathology. To observe this, we first created a subacute injury model of sufficient duration to allow inflammatory cell recruitment. Mice were injected intraperitoneally with an anti-GD1b monoclonal antibody that binds strongly to mouse motor nerve axons. Subsequently, mice received normal human serum as a source of complement. Dosing was titrated to allow humane survival of mice over a period of 3 days, yet still induce the characteristic neurological impairment. Behaviour and pathology were assessed in vivo using whole-body plethysmography and post-sacrifice by immunofluorescence and flow cytometry. ex vivo nerve-muscle preparations were used to investigate the acute phagocytic role of pSCs following distal nerve injury. Following complement activation at distal intramuscular nerve sites in the diaphragm macrophage localisation or numbers are not altered, nor do they shift to a pro- or anti-inflammatory phenotype. Similarly, neutrophils are not significantly recruited. Instead, ex vivo nerve-muscle preparations exposed to AGAb plus complement reveal that pSCs rapidly become phagocytic and engulf axonal debris. These data suggest that pSCs, rather than inflammatory cells, are the major cellular vehicle for axonal debris clearance following distal nerve injury, in contrast to larger nerve bundles where macrophage-mediated clearance predominates.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Gangliósidos/inmunología , Síndrome de Guillain-Barré , Neuronas Motoras , Unión Neuromuscular , Fagocitosis/fisiología , Terminales Presinápticos , Células de Schwann/fisiología , Animales , Anticuerpos Monoclonales/administración & dosificación , Conducta Animal/fisiología , Activación de Complemento/inmunología , Modelos Animales de Enfermedad , Femenino , Síndrome de Guillain-Barré/inmunología , Síndrome de Guillain-Barré/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Neuronas Motoras/inmunología , Neuronas Motoras/patología , Unión Neuromuscular/inmunología , Unión Neuromuscular/patología , Terminales Presinápticos/inmunología , Terminales Presinápticos/patología
7.
J Infect Dis ; 218(2): 324-335, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29236994

RESUMEN

Acute rheumatic fever and rheumatic heart disease (ARF/RHD) have long been described as autoimmune sequelae of Streptococcus pyogenes or group A streptococcal (GAS) infection. Both antibody and T-cell responses against immunodominant GAS virulence factors, including M protein, cross-react with host tissue proteins, triggering an inflammatory response leading to permanent heart damage. However, in some ARF/RHD-endemic regions, throat carriage of GAS is low. Because Streptococcus dysgalactiae subspecies equisimilis organisms, also known as ß-hemolytic group C streptococci and group G streptococci (GGS), also express M protein, we postulated that streptococci other than GAS may have the potential to initiate or exacerbate ARF/RHD. Using a model initially developed to investigate the uniquely human disease of ARF/RHD, we have discovered that GGS causes interleukin 17A/interferon γ-induced myocarditis and valvulitis, hallmarks of ARF/RHD. Remarkably the histological, immunological, and functional changes in the hearts of rats exposed to GGS are identical to those exposed to GAS. Furthermore, antibody cross-reactivity to cardiac myosin was comparable in both GGS- and GAS-exposed animals, providing additional evidence that GGS can induce and/or exacerbate ARF/RHD.


Asunto(s)
Enfermedades Autoinmunes/etiología , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Cardiopatía Reumática/etiología , Infecciones Estreptocócicas/patología , Streptococcus/inmunología , Animales , Antígenos Bacterianos/inmunología , Enfermedades Autoinmunes/microbiología , Enfermedades Autoinmunes/fisiopatología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Modelos Animales de Enfermedad , Femenino , Enfermedades de las Válvulas Cardíacas/etiología , Enfermedades de las Válvulas Cardíacas/microbiología , Enfermedades de las Válvulas Cardíacas/fisiopatología , Miocarditis/etiología , Miocarditis/microbiología , Miocarditis/fisiopatología , Ratas Endogámicas Lew , Cardiopatía Reumática/microbiología , Cardiopatía Reumática/fisiopatología , Streptococcus/patogenicidad
8.
Brain ; 139(Pt 6): 1657-65, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27017187

RESUMEN

SEE VAN DOORN AND JACOBS DOI101093/BRAIN/AWW078 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE : In axonal forms of Guillain-Barré syndrome, anti-ganglioside antibodies bind gangliosides on nerve surfaces, thereby causing injury through complement activation and immune cell recruitment. Why some nerve regions are more vulnerable than others is unknown. One reason may be that neuronal membranes with high endocytic activity, including nerve terminals involved in neurotransmitter recycling, are able to endocytose anti-ganglioside antibodies from the cell surface so rapidly that antibody-mediated injury is attenuated. Herein we investigated whether endocytic clearance of anti-ganglioside antibodies by nerve terminals might also be of sufficient magnitude to deplete circulating antibody levels. Remarkably, systemically delivered anti-ganglioside antibody in mice was so avidly cleared from the circulation by endocytosis at ganglioside-expressing plasma membranes that it was rapidly rendered undetectable in serum. A major component of the clearance occurred at motor nerve terminals of neuromuscular junctions, from where anti-ganglioside antibody was retrogradely transported to the motor neuron cell body in the spinal cord, recycled to the plasma membrane, and secreted into the surrounding spinal cord. Uptake at the neuromuscular junction represents a major unexpected pathway by which pathogenic anti-ganglioside antibodies, and potentially other ganglioside binding proteins, are cleared from the systemic circulation and also covertly delivered to the central nervous system.


Asunto(s)
Anticuerpos/metabolismo , Endocitosis/inmunología , Gangliósidos/inmunología , Terminales Presinápticos/metabolismo , Animales , Anticuerpos/sangre , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , N-Acetilgalactosaminiltransferasas/genética , Unión Neuromuscular/metabolismo , Polipéptido N-Acetilgalactosaminiltransferasa
9.
J Card Fail ; 22(7): 520-8, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26941007

RESUMEN

BACKGROUND: Host autoimmune activity in myocarditis has been proposed to play a role in development of cardiac disease, but evidence of autoimmunity and relationship to outcomes have not been evaluated in pediatric myocarditis. METHODS: We performed a multi-institutional study of children with clinical myocarditis. Newly diagnosed patients were followed for up to 12 months and previously diagnosed patients at a single follow-up for serum levels of autoantibodies to human cardiac myosin, beta-adrenergic receptors 1 and 2, muscarinic-2 receptors, and antibody-mediated protein kinase A (PKA) activation in heart cells in culture. Results were compared with those of healthy control children. RESULTS: Both previously diagnosed patient at follow-up (P = .0061) and newly diagnosed patients at presentation (P = .0127) had elevated cardiac myosin antibodies compared with control subjects. Antibody levels were not associated with recovery status at follow-up in either group. PKA activation was higher at presentation in the newly diagnosed patients who did not recovery normal function (P = .042). CONCLUSIONS: Children with myocarditis have evidence of autoantibodies against human cardiac myosin at diagnosis and follow-up compared with control subjects. Differences in antibody-mediated cell signaling may contribute to differences in patient outcomes, as suggested by elevated antibody-mediated PKA activation in heart cells by the serum from nonrecovered patients.


Asunto(s)
Autoanticuerpos/inmunología , Autoinmunidad , Miosinas Cardíacas/inmunología , Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Miocarditis/inmunología , Miocitos Cardíacos/inmunología , Autoanticuerpos/sangre , Niño , Preescolar , Proteínas Quinasas Dependientes de AMP Cíclico/análisis , Corazón/diagnóstico por imagen , Corazón/fisiopatología , Humanos , Lactante , Miocarditis/sangre , Miocarditis/diagnóstico por imagen , Miocitos Cardíacos/química
10.
J Immunol ; 192(9): 4074-82, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24670798

RESUMEN

Structural congenital heart disease (CHD) has not previously been linked to autoimmunity. In our study, we developed an autoimmune model of structural CHD that resembles hypoplastic left heart syndrome (HLHS), a life-threatening CHD primarily affecting the left ventricle. Because cardiac myosin (CM) is a dominant autoantigen in autoimmune heart disease, we hypothesized that immunization with CM might lead to transplacental passage of maternal autoantibodies and a prenatal HLHS phenotype in exposed fetuses. Elevated anti-CM autoantibodies in maternal and fetal sera, as well as IgG reactivity in fetal myocardium, were correlated with structural CHD that included diminished left ventricular cavity dimensions in the affected progeny. Further, fetuses that developed a marked HLHS phenotype had elevated serum titers of anti-ß-adrenergic receptor Abs, as well as increased protein kinase A activity, suggesting a potential mechanism for the observed pathological changes. Our maternal-fetal model presents a new concept linking autoimmunity against CM and cardiomyocyte proliferation with cardinal features of HLHS. To our knowledge, this report shows the first evidence in support of a novel immune-mediated mechanism for pathogenesis of structural CHD that may have implications in its future diagnosis and treatment.


Asunto(s)
Autoinmunidad/inmunología , Miosinas Cardíacas/inmunología , Síndrome del Corazón Izquierdo Hipoplásico/inmunología , Animales , Autoanticuerpos/inmunología , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Cardiopatías Congénitas/inmunología , Cardiopatías Congénitas/patología , Síndrome del Corazón Izquierdo Hipoplásico/patología , Inmunohistoquímica , Ratas , Ratas Endogámicas Lew
11.
J Neurosci ; 34(3): 880-91, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24431446

RESUMEN

Gangliosides are widely expressed sialylated glycosphingolipids with multifunctional properties in different cell types and organs. In the nervous system, they are highly enriched in both glial and neuronal membranes. Mice lacking complex gangliosides attributable to targeted ablation of the B4galnt1 gene that encodes ß-1,4-N-acetylegalactosaminyltransferase 1 (GalNAc-transferase; GalNAcT(-/-)) develop normally before exhibiting an age-dependent neurodegenerative phenotype characterized by marked behavioral abnormalities, central and peripheral axonal degeneration, reduced myelin volume, and loss of axo-glial junction integrity. The cell biological substrates underlying this neurodegeneration and the relative contribution of either glial or neuronal gangliosides to the process are unknown. To address this, we generated neuron-specific and glial-specific GalNAcT rescue mice crossed on the global GalNAcT(-/-) background [GalNAcT(-/-)-Tg(neuronal) and GalNAcT(-/-)-Tg(glial)] and analyzed their behavioral, morphological, and electrophysiological phenotype. Complex gangliosides, as assessed by thin-layer chromatography, mass spectrometry, GalNAcT enzyme activity, and anti-ganglioside antibody (AgAb) immunohistology, were restored in both neuronal and glial GalNAcT rescue mice. Behaviorally, GalNAcT(-/-)-Tg(neuronal) retained a normal "wild-type" (WT) phenotype throughout life, whereas GalNAcT(-/-)-Tg(glial) resembled GalNAcT(-/-) mice, exhibiting progressive tremor, weakness, and ataxia with aging. Quantitative electron microscopy demonstrated that GalNAcT(-/-) and GalNAcT(-/-)-Tg(glial) nerves had significantly increased rates of axon degeneration and reduced myelin volume, whereas GalNAcT(-/-)-Tg(neuronal) and WT appeared normal. The increased invasion of the paranode with juxtaparanodal Kv1.1, characteristically seen in GalNAcT(-/-) and attributed to a breakdown of the axo-glial junction, was normalized in GalNAcT(-/-)-Tg(neuronal) but remained present in GalNAcT(-/-)-Tg(glial) mice. These results indicate that neuronal rather than glial gangliosides are critical to the age-related maintenance of nervous system integrity.


Asunto(s)
Envejecimiento/metabolismo , Gangliósidos/deficiencia , Regulación Enzimológica de la Expresión Génica , N-Acetilgalactosaminiltransferasas/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Fenotipo , Envejecimiento/genética , Envejecimiento/patología , Animales , Axones/metabolismo , Axones/patología , Gangliósidos/genética , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , N-Acetilgalactosaminiltransferasas/biosíntesis , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Polipéptido N-Acetilgalactosaminiltransferasa
12.
J Immunol ; 191(11): 5524-41, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24184556

RESUMEN

How autoantibodies target the brain and lead to disease in disorders such as Sydenham chorea (SC) is not known. SC is characterized by autoantibodies against the brain and is the main neurologic manifestation of streptococcal-induced rheumatic fever. Previously, our novel SC-derived mAb 24.3.1 was found to recognize streptococcal and brain Ags. To investigate in vivo targets of human mAb 24.3.1, VH/VL genes were expressed in B cells of transgenic (Tg) mice as functional chimeric human VH 24.3.1-mouse C-region IgG1(a) autoantibody. Chimeric human-mouse IgG1(a) autoantibody colocalized with tyrosine hydroxylase in the basal ganglia within dopaminergic neurons in vivo in VH 24.3.1 Tg mice. Both human mAb 24.3.1 and IgG1(a) in Tg sera were found to react with human dopamine D2 receptor (D2R). Reactivity of chorea-derived mAb 24.3.1 or SC IgG with D2R was confirmed by dose-dependent inhibitory signaling of D2R as a potential consequence of targeting dopaminergic neurons, reaction with surface-exposed FLAG epitope-tagged D2R, and blocking of Ab reactivity by an extracellular D2R peptide. IgG from SC and a related subset of streptococcal-associated behavioral disorders called "pediatric autoimmune neuropsychiatric disorder associated with streptococci" (PANDAS) with small choreiform movements reacted in ELISA with D2R. Reaction with FLAG-tagged D2R distinguished SC from PANDAS, whereas sera from both SC and PANDAS induced inhibitory signaling of D2R on transfected cells comparably to dopamine. In this study, we define a mechanism by which the brain may be altered by Ab in movement and behavioral disorders.


Asunto(s)
Corea/inmunología , Neuronas Dopaminérgicas/metabolismo , Receptores de Dopamina D2/metabolismo , Fiebre Reumática/inmunología , Infecciones Estreptocócicas/inmunología , Animales , Antígenos Bacterianos/inmunología , Autoanticuerpos/genética , Autoanticuerpos/metabolismo , Ganglios Basales/patología , Niño , Corea/etiología , Reacciones Cruzadas , Dopamina/metabolismo , Neuronas Dopaminérgicas/inmunología , Gangliósido G(M1)/análogos & derivados , Gangliósido G(M1)/inmunología , Células HEK293 , Humanos , Inmunoglobulina G/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Dopamina D2/genética , Proteínas Recombinantes de Fusión/genética , Fiebre Reumática/etiología , Transducción de Señal , Infecciones Estreptocócicas/complicaciones , Transgenes/genética
14.
J Biol Chem ; 288(42): 30734-30741, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24043632

RESUMEN

Functional autoantibodies to the autonomic receptors are increasingly recognized in the pathophysiology of cardiovascular diseases. To date, no human activating monoclonal autoantibodies to these receptors have been available. In this study, we describe for the first time a ß2-adrenergic receptor (ß2AR)-activating monoclonal autoantibody (C5F2) produced from the lymphocytes of a patient with idiopathic postural hypotension. C5F2, an IgG3 isotype, recognizes an epitope in the N terminus of the second extracellular loop (ECL2) of ß2AR. Surface plasmon resonance analysis revealed high binding affinity for the ß2AR ECL2 peptide. Immunoblotting and immunofluorescence demonstrated specific binding to ß2AR in H9c2 cardiomyocytes, CHO cells expressing human ß2AR, and rat aorta. C5F2 stimulated cyclic AMP production in ß2AR-transfected CHO cells and induced potent dilation of isolated rat cremaster arterioles, both of which were specifically blocked by the ß2AR-selective antagonist ICI-118551 and by the ß2AR ECL2 peptide. This monoclonal antibody demonstrated sufficient activity to produce postural hypotension in its host. Its availability provides a unique opportunity to identify previously unrecognized causes and new pharmacological management of postural hypotension and other cardiovascular diseases.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Autoanticuerpos/inmunología , Hipotensión Ortostática/inmunología , Hipotensión Ortostática/fisiopatología , Inmunoglobulina G/inmunología , Receptores Adrenérgicos beta 2/inmunología , Vasodilatadores/inmunología , Antagonistas Adrenérgicos beta/farmacología , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/farmacología , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Aorta/fisiopatología , Arteriolas/metabolismo , Arteriolas/patología , Arteriolas/fisiopatología , Autoanticuerpos/sangre , Autoanticuerpos/farmacología , Células CHO , Cricetinae , Cricetulus , Humanos , Hipotensión Ortostática/sangre , Hipotensión Ortostática/genética , Hipotensión Ortostática/patología , Inmunoglobulina G/sangre , Inmunoglobulina G/farmacología , Masculino , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Péptidos/inmunología , Péptidos/farmacología , Propanolaminas/farmacología , Ratas , Receptores Adrenérgicos beta 2/sangre , Receptores Adrenérgicos beta 2/genética , Resonancia por Plasmón de Superficie , Vasodilatadores/sangre , Vasodilatadores/farmacología
15.
Am J Physiol Heart Circ Physiol ; 306(3): H422-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24271491

RESUMEN

Previous studies demonstrated burst pacing and intravenous infusion of ACh induced sustained atrial tachycardia when rabbits were immunized to produce ß2-adrenergic receptor (ß2AR)-activating autoantibodies. The objective of this study was to examine the arrhythmogenic effect of ß1-adrenergic receptor (ß1AR)-activating autoantibodies in the rabbit. Eight New Zealand white rabbits were immunized with a ß1AR second extracellular loop peptide to raise ß1AR antibody titers. A catheter-based electrophysiological study was performed on anesthetized rabbits before and after immunization. Arrhythmia occurrence was determined in response to burst pacing before and after ACh infusion in incremental concentrations of 10 µM, 100 µM, and 1 mM. The baseline sinus heart rate before and after immunization averaged 149 ± 17 per min and 169 ± 16 per min, respectively (P < 0.05). In the preimmune studies, there were five sustained (≥10 s) arrhythmias in 32 induction attempts, which occurred in only four of eight rabbits. In the postimmune studies, there were 22 sustained arrhythmias in 32 induction attempts, which occurred in all eight rabbits (P < 0.0001 for the independent effect of immunization). Of the 22 sustained arrhythmias postimmunization, 15 were sinus tachycardia compared with only two before immunization (P < 0.01 for the independent effect of immunization). Postimmune (but not preimmune) rabbit sera demonstrated specific binding to ß1AR and induced significant ß1AR activation in transfected cells in vitro. No cross-reactivity with ß2AR was observed. In conclusion, in contrast with rabbits with ß2AR-activating autoantibodies that demonstrate predominantly atrial tachycardias, enhanced autoantibody activation of ß1AR in the rabbit leads to tachyarrhythmias mainly in the form of sustained sinus tachycardia.


Asunto(s)
Arritmias Cardíacas/inmunología , Autoanticuerpos/metabolismo , Receptores Adrenérgicos beta 1/inmunología , Acetilcolina/farmacología , Animales , Arritmias Cardíacas/metabolismo , Autoanticuerpos/inmunología , Frecuencia Cardíaca , Conejos , Receptores Adrenérgicos beta 1/metabolismo
16.
Brain Behav Immun ; 38: 249-62, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24561489

RESUMEN

Group A ß-hemolytic streptococcal (GAS) infection is associated with a spectrum of neuropsychiatric disorders. The leading hypothesis regarding this association proposes that a GAS infection induces the production of auto-antibodies, which cross-react with neuronal determinants in the brain through the process of molecular mimicry. We have recently shown that exposure of rats to GAS antigen leads to the production of anti-neuronal antibodies concomitant with the development of behavioral alterations. The present study tested the causal role of the antibodies by assessing the behavior of naïve rats following passive transfer of purified antibodies from GAS-exposed rats. Immunoglobulin G (IgG) purified from the sera of GAS-exposed rats was infused directly into the striatum of naïve rats over a 21-day period. Their behavior in the induced-grooming, marble burying, food manipulation and beam walking assays was compared to that of naïve rats infused with IgG purified from adjuvant-exposed rats as well as of naïve rats. The pattern of in vivo antibody deposition in rat brain was evaluated using immunofluorescence and colocalization. Infusion of IgG from GAS-exposed rats to naïve rats led to behavioral and motor alterations partially mimicking those seen in GAS-exposed rats. IgG from GAS-exposed rats reacted with D1 and D2 dopamine receptors and 5HT-2A and 5HT-2C serotonin receptors in vitro. In vivo, IgG deposits in the striatum of infused rats colocalized with specific brain proteins such as dopamine receptors, the serotonin transporter and other neuronal proteins. Our results demonstrate the potential pathogenic role of autoantibodies produced following exposure to GAS in the induction of behavioral and motor alterations, and support a causal role for autoantibodies in GAS-related neuropsychiatric disorders.


Asunto(s)
Cuerpo Estriado/inmunología , Inmunoglobulina G/inmunología , Streptococcus pyogenes/inmunología , Animales , Conducta Animal , Cuerpo Estriado/metabolismo , Masculino , Actividad Motora , Ratas , Ratas Endogámicas Lew , Receptores Dopaminérgicos/metabolismo , Receptores de Serotonina/metabolismo
17.
Infect Dis Obstet Gynecol ; 2014: 639141, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25210420

RESUMEN

OBJECTIVES: To report the pharyngeal colonization rate of ß-hemolytic streptococci and changes in the value of antistreptolysin O (ASO) and anti-DNase B serology titers during pregnancy. METHODS: Healthy pregnant women were recruited and blood was drawn in each trimester. The upper limit of normal (ULN) values for ASO and anti-DNase B was calculated for each trimester. Throat swabs were collected for culture and positive cultures were further assessed for the identification of serogroup of the isolated ß-hemolytic streptococcus. RESULTS: Out of a total of 126 pregnant women, 34.1% had positive throat cultures. Group C and group G strains were isolated in 18.2% of throat cultures while group F was detected in 13.5% of cases. The rate of colonization with GAS was 1.6%. There was an overall drop in ASO titer during pregnancy while anti-DNase B titers remained relatively unchanged. ULN values of 164(IU), 157(IU), and 156(IU) were calculated for ASO at the first, second, and third trimesters, respectively. Based on the ULN values, 28.6% of patients had recent streptococcal exposure. CONCLUSIONS: These results show that pregnant women act as a reservoir for spreading potentially immunogenic (groups C and G) and disease producing (group F) virulent strains of streptococci.


Asunto(s)
Portador Sano/microbiología , Faringitis/microbiología , Complicaciones Infecciosas del Embarazo/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/aislamiento & purificación , Adulto , Portador Sano/epidemiología , Femenino , Humanos , Faringitis/epidemiología , Faringe/microbiología , Embarazo , Complicaciones Infecciosas del Embarazo/epidemiología , Infecciones Estreptocócicas/epidemiología , Estados Unidos/epidemiología , Adulto Joven
18.
Synapse ; 67(7): 382-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23401234

RESUMEN

Gangliosides are glycosphingolipids highly enriched in neural plasma membranes, where they mediate a diverse range of functions and can act as targets for auto-antibodies present in human immune-mediated neuropathy sera. The ensuing autoimmune injury results in axonal and motor nerve terminal (mNT) degeneration. Both aging and ganglioside-deficiency have been linked to impaired axonal regeneration. To assess the effects of age and ganglioside expression on mNT regeneration in an autoimmune injury paradigm, anti-ganglioside antibodies and complement were applied to young adult and aged mice wildtype (WT) mice, mice deficient in either b- and c-series (GD3sKO) or mice deficient in all complex gangliosides (GM2sKO). The extent of mNT injury and regeneration was assessed immediately or after 5 days, respectively. Depending on ganglioside expression and antibody-specificity, either a selective mNT injury or a combined injury of mNTs and neuromuscular glial cells was elicited. Immediately after induction of the injury, between 1.5% and 11.8% of neuromuscular junctions (NMJs) in the young adult groups exhibited healthy mNTs. Five days later, most NMJs, regardless of age and strain, had recovered their mNTs. No significant differences could be observed between young and aged WT and GM2sKO mice; aged GD3sKO showed a mildly impaired rate of mNT regeneration when compared with their younger counterparts. Comparable rates were observed between all strains in the young and the aged mice. In summary, the rate of mNT regeneration following anti-ganglioside antibody and complement-mediated injury does not differ majorly between young adult and aged mice irrespective of the expression of particular gangliosides.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Gangliósido G(M2)/metabolismo , Gangliósidos/metabolismo , Regeneración Nerviosa/inmunología , Unión Neuromuscular/inmunología , Factores de Edad , Animales , Proteínas del Sistema Complemento/inmunología , Gangliósido G(M2)/inmunología , Gangliósidos/inmunología , Ratones , Ratones Noqueados , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/metabolismo , Unión Neuromuscular/fisiopatología , Células de Schwann/metabolismo , Células de Schwann/fisiología , Sialiltransferasas/genética , Sialiltransferasas/metabolismo
19.
J Immunol ; 187(2): 1023-30, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677143

RESUMEN

Chronic allograft vasculopathy (CAV) contributes to heart transplant failure, yet its pathogenesis is incompletely understood. Although cellular and humoral alloimmunity are accepted pathogenic mediators, animal models suggest that T cells and Abs reactive to graft-expressed autoantigens, including cardiac myosin (CM), could participate. To test the relationship between CAV and anti-CM autoimmunity in humans, we performed a cross-sectional study of 72 heart transplant recipients: 40 with CAV and 32 without. Sera from 65% of patients with CAV contained anti-CM Abs, whereas <10% contained Abs to other autoantigens (p < 0.05), and only 18% contained anti-HLA Abs (p < 0.05 versus anti-CM). In contrast, 13% of sera from patients without CAV contained anti-CM Abs (p < 0.05; odds ratio [OR], associating CAV with anti-CM Ab = 13, 95% confidence interval [CI] 3.79-44.6). Multivariable analysis confirmed the association to be independent of time posttransplant and the presence of anti-HLA Abs (OR = 28, 95% CI 5.77-133.56). PBMCs from patients with CAV responded more frequently to, and to a broader array of, CM-derived peptides than those without CAV (p = 0.01). Detection of either CM-peptide-reactive T cells or anti-CM Abs was highly and independently indicative of CAV (OR = 45, 95% CI 4.04-500.69). Our data suggest detection of anti-CM immunity could be used as a biomarker for outcome in heart transplantation recipients and support the need for further studies to assess whether anti-CM immunity is a pathogenic mediator of CAV.


Asunto(s)
Miosinas Cardíacas/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Trasplante de Corazón/inmunología , Trasplante de Corazón/patología , Anciano , Secuencia de Aminoácidos , Animales , Autoanticuerpos/biosíntesis , Biomarcadores/sangre , Enfermedad Crónica , Complemento C4b/inmunología , Estudios Transversales , Endotelio Vascular/metabolismo , Femenino , Rechazo de Injerto/diagnóstico , Antígenos HLA/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Infiltración Neutrófila/inmunología , Fragmentos de Péptidos/inmunología , Proyectos Piloto , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
20.
Psychiatr Clin North Am ; 46(1): 69-88, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36740356

RESUMEN

Sydenham chorea (SC), pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections (PANDAS) and pediatric acute-onset neuropsychiatric syndrome (PANS) are postinfectious neuroinflammatory diseases that involve the basal ganglia and have obsessive-compulsive disorder as a major manifestation. As is true for many childhood rheumatological diseases and neuroinflammatory diseases, SC, PANDAS and PANS lack clinically available, rigorous diagnostic biomarkers and randomized clinical trials. Research on the treatment of these disorders depend on three complementary modes of intervention including: treating the symptoms, treating the source of inflammation, and treating disturbances of the immune system. Future studies should aim to integrate neuroimaging, inflammation, immunogenetic, and clinical data (noting the stage in the clinical course) to increase our understanding and treatment of SC, PANDAS, PANS, and all other postinfectious/immune-mediated behavioral disorders.


Asunto(s)
Enfermedades Autoinmunes , Corea , Trastorno Obsesivo Compulsivo , Infecciones Estreptocócicas , Niño , Humanos , Enfermedades Neuroinflamatorias , Corea/complicaciones , Corea/diagnóstico , Enfermedades Autoinmunes/complicaciones , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/tratamiento farmacológico , Trastorno Obsesivo Compulsivo/complicaciones , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/diagnóstico , Infecciones Estreptocócicas/tratamiento farmacológico , Inflamación/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA