Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Brain ; 147(3): 839-848, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-38123517

RESUMEN

Intrathecal IgM production in multiple sclerosis is associated with a worse disease course. To investigate pathogenic relevance of autoreactive IgM in multiple sclerosis, CSF from two independent cohorts, including multiple sclerosis patients and controls, were screened for antibody binding to induced pluripotent stem cell-derived neurons and astrocytes, and a panel of CNS-related cell lines. IgM binding to a primitive neuro-ectodermal tumour cell line discriminated 10% of multiple sclerosis donors from controls. Transcriptomes of single IgM producing CSF B cells from patients with cell-binding IgM were sequenced and used to produce recombinant monoclonal antibodies for characterization and antigen identification. We produced five cell-binding recombinant IgM antibodies, of which one, cloned from an HLA-DR + plasma-like B cell, mediated antigen-dependent complement activation. Immunoprecipitation and mass spectrometry, and biochemical and transcriptome analysis of the target cells identified the iron transport scavenger protein SCARA5 as the antigen target of this antibody. Intrathecal injection of a SCARA5 antibody led to an increased T cell infiltration in an experimental autoimmune encephalomyelitis (EAE) model. CSF IgM might contribute to CNS inflammation in multiple sclerosis by binding to cell surface antigens like SCARA5 and activating complement, or by facilitating immune cell migration into the brain.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Inmunoglobulina M , Esclerosis Múltiple , Receptores Depuradores de Clase A , Animales , Humanos , Anticuerpos Monoclonales , Línea Celular Tumoral , Inmunoglobulina M/líquido cefalorraquídeo , Proteínas de Transporte de Membrana , Esclerosis Múltiple/líquido cefalorraquídeo , Esclerosis Múltiple/inmunología , Receptores Depuradores de Clase A/inmunología
2.
Proc Natl Acad Sci U S A ; 119(31): e2205042119, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35881799

RESUMEN

Dimethyl fumarate (DMF) is an immunomodulatory treatment for multiple sclerosis (MS). Despite its wide clinical use, the mechanisms underlying clinical response are not understood. This study aimed to reveal immune markers of therapeutic response to DMF treatment in MS. For this purpose, we prospectively collected peripheral blood mononuclear cells (PBMCs) from a highly characterized cohort of 44 individuals with MS before and at 12 and 48 wk of DMF treatment. Single cells were profiled using high-dimensional mass cytometry. To capture the heterogeneity of different immune subsets, we adopted a bioinformatic multipanel approach that allowed cell population-cluster assignment of more than 50 different parameters, including lineage and activation markers as well as chemokine receptors and cytokines. Data were further analyzed in a semiunbiased fashion implementing a supervised representation learning approach to capture subtle longitudinal immune changes characteristic for therapy response. With this approach, we identified a population of memory T helper cells expressing high levels of neuroinflammatory cytokines (granulocyte-macrophage colony-stimulating factor [GM-CSF], interferon γ [IFNγ]) as well as CXCR3, whose abundance correlated with treatment response. Using spectral flow cytometry, we confirmed these findings in a second cohort of patients. Serum neurofilament light-chain levels confirmed the correlation of this immune cell signature with axonal damage. The identified cell population is expanded in peripheral blood under natalizumab treatment, substantiating a specific role in treatment response. We propose that depletion of GM-CSF-, IFNγ-, and CXCR3-expressing T helper cells is the main mechanism of action of DMF and allows monitoring of treatment response.


Asunto(s)
Biomarcadores Farmacológicos , Citocinas , Dimetilfumarato , Inmunosupresores , Esclerosis Múltiple , Linfocitos T Colaboradores-Inductores , Biomarcadores Farmacológicos/metabolismo , Citocinas/metabolismo , Dimetilfumarato/farmacología , Dimetilfumarato/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Interferón gamma/metabolismo , Depleción Linfocítica , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Análisis de la Célula Individual , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología
3.
EMBO Rep ; 23(7): e53956, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35548920

RESUMEN

To investigate the class-dependent properties of anti-viral IgM antibodies, we use membrane antigen capture activated cell sorting to isolate spike-protein-specific B cells from donors recently infected with SARS-CoV-2, allowing production of recombinant antibodies. We isolate 20, spike-protein-specific antibodies of classes IgM, IgG, and IgA, none of which shows any antigen-independent binding to human cells. Two antibodies of class IgM mediate virus neutralization at picomolar concentrations, but this potency is lost following artificial switch to IgG. Although, as expected, the IgG versions of the antibodies appear to have lower avidity than their IgM parents, this is not sufficient to explain the loss of potency.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anticuerpos Monoclonales , Anticuerpos Antivirales , Humanos , Inmunoglobulina G , Inmunoglobulina M
4.
Acta Neuropathol ; 144(5): 1005-1025, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36074148

RESUMEN

Myasthenia gravis is an autoimmune disorder defined by muscle weakness and fatigability associated with antibodies against proteins of the neuromuscular junction (NMJ). The most common autoantibody target is the acetylcholine receptor (AChR). Three mechanisms have been postulated by which autoantibodies might interfere with neurotransmission: direct antagonism of the receptor, complement-mediated destruction of the postsynaptic membrane, and enhanced internalization of the receptor. It is very likely that more than one of these mechanisms act in parallel. Dissecting the mechanisms of autoantibody-mediated pathology requires patient-derived, monoclonal antibodies. Using membrane antigen capture activated cell sorting (MACACS), we isolated AChR-specific B cells from patients with myasthenia gravis, and produced six recombinant antibodies. All AChR-specific antibodies were hypermutated, including isotypes IgG1, IgG3, and IgG4, and recognized different subunits of the AChR. Despite clear binding, none of the individual antibodies showed significant antagonism of the AChR measured in an in vitro neuromuscular synapse model, or AChR-dependent complement activation, and they did not induce myasthenic signs in vivo. However, combinations of antibodies induced strong complement activation in vitro, and severe weakness in a passive transfer myasthenia gravis rat model, associated with NMJ destruction and complement activation in muscle. The strongest complement activation was mediated by combinations of antibodies targeting disparate subunits of the AChR, and such combinations also induced the formation of large clusters of AChR on the surface of live cells in vitro. We propose that synergy between antibodies of different epitope specificities is a fundamental feature of this disease, and possibly a general feature of complement-mediated autoimmune diseases. The importance of synergistic interaction between antibodies targeting different subunits of the receptor can explain the well-known discrepancy between serum anti-AChR titers and clinical severity, and has implications for therapeutic strategies currently under investigation.


Asunto(s)
Miastenia Gravis , Animales , Anticuerpos Monoclonales , Autoanticuerpos , Análisis por Conglomerados , Activación de Complemento , Proteínas del Sistema Complemento , Epítopos , Inmunoglobulina G/metabolismo , Miastenia Gravis/patología , Ratas , Receptores Colinérgicos , Receptores de Complemento
5.
Proc Natl Acad Sci U S A ; 114(4): 734-739, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28057865

RESUMEN

Autoantibodies against myelin oligodendrocyte glycoprotein (MOG) are associated with autoimmune central nervous system diseases like acute disseminated encephalomyelitis (ADEM). For ADEM, it is speculated that a preceding infection is the trigger of the autoimmune response, but the mechanism connecting the infection to the production of MOG antibodies remains a mystery. We reasoned that the ability of B cells to capture cognate antigen from cell membranes, along with small quantities of coexpressed "bystander" antigens, might enable B-cell escape from tolerance. We tested this hypothesis using influenza hemagglutinin as a model viral antigen and transgenic, MOG-specific B cells. Using flow cytometry and live and fixed cell microscopy, we show that MOG-specific B cells take up large amounts of MOG from cell membranes. Uptake of the antigen from the membrane leads to a strong activation of the capturing B cell. When influenza hemagglutinin is also present in the membrane of the target cell, it can be cocaptured with MOG by MOG-specific B cells via the B-cell receptor. Hemagglutinin and MOG are both presented to T cells, which in turn are activated and proliferate. As a consequence, MOG-specific B cells get help from hemagglutinin-specific T cells to produce anti-MOG antibodies. In vivo, the transfer of MOG-specific B cells into recipient mice after the cocapture of MOG and hemagglutinin leads to the production of class-switched anti-MOG antibodies, dependent on the presence of hemagglutinin-specific T cells. This mechanism offers a link between infection and autoimmunity.


Asunto(s)
Antígenos Virales/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Animales , Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Línea Celular , Membrana Celular/inmunología , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Linfocitos T/inmunología
6.
J Neuroinflammation ; 12: 46, 2015 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-25889963

RESUMEN

BACKGROUND: Antibodies against myelin oligodendrocyte glycoprotein (MOG) have been identified in a subgroup of pediatric patients with inflammatory demyelinating disease of the central nervous system (CNS) and in some patients with neuromyelitis optica spectrum disorder (NMOSD). The aim of this study was to examine the frequency, clinical features, and long-term disease course of patients with anti-MOG antibodies in a European cohort of NMO/NMOSD. FINDINGS: Sera from 48 patients with NMO/NMOSD and 48 patients with relapsing-remitting multiple sclerosis (RR-MS) were tested for anti-aquaporin-4 (AQP4) and anti-MOG antibodies with a cell-based assay. Anti-MOG antibodies were found in 4/17 patients with AQP4-seronegative NMO/NMOSD, but in none of the AQP4-seropositive NMO/NMOSD (n = 31) or RR-MS patients (n = 48). MOG-seropositive patients tended towards younger disease onset with a higher percentage of patients with pediatric (<18 years) disease onset (MOG+, AQP4+, MOG-/AQP4-: 2/4, 3/31, 0/13). MOG-seropositive patients presented more often with positive oligoclonal bands (OCBs) (3/3, 5/29, 1/13) and brain magnetic resonance imaging (MRI) lesions during disease course (2/4, 5/31, 1/13). Notably, the mean time to the second attack affecting a different CNS region was longer in the anti-MOG antibody-positive group (11.3, 3.2, 3.4 years). CONCLUSIONS: MOG-seropositive patients show a diverse clinical phenotype with clinical features resembling both NMO (attacks mainly confined to the spinal cord and optic nerves) and MS with an opticospinal presentation (positive OCBs, brain lesions). Anti-MOG antibodies can serve as a diagnostic and maybe prognostic tool in patients with an AQP4-seronegative NMO phenotype and should be tested in those patients.


Asunto(s)
Autoanticuerpos/sangre , Glicoproteína Mielina-Oligodendrócito/inmunología , Neuromielitis Óptica/sangre , Adulto , Anciano , Acuaporina 4/inmunología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/sangre , Estudios Retrospectivos , Adulto Joven
7.
Int J Mol Sci ; 16(7): 16576-92, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26197319

RESUMEN

While over the past decades T cells have been considered key players in the pathogenesis of multiple sclerosis (MS), it has only recently become evident that B cells have a major contributing role. Our understanding of the role of B cells has evolved substantially following the clinical success of B cell-targeting therapies and increasing experimental evidence for significant B cell involvement. Rather than mere antibody-producing cells, it is becoming clear that they are team players with the capacity to prime and regulate T cells, and function both as pro- and anti-inflammatory mediators. However, despite tremendous efforts, the target antigen(s) of B cells in MS have yet to be identified. The first part of this review summarizes the clinical evidence and results from animal studies pointing to the relevance of B cells in the pathogenesis of MS. The second part gives an overview of the currently known potential autoantigen targets. The third part recapitulates and critically appraises the currently available B cell-directed therapies.


Asunto(s)
Autoanticuerpos/inmunología , Linfocitos B/inmunología , Esclerosis Múltiple/inmunología , Animales , Humanos , Inmunoterapia/métodos , Esclerosis Múltiple/etiología , Esclerosis Múltiple/terapia , Medicina de Precisión/métodos
9.
Proc Natl Acad Sci U S A ; 107(32): 14443-8, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660723

RESUMEN

Soluble antigens diffuse out of the brain and can thus stimulate a systemic immune response, whereas particulate antigens (from infectious agents or tumor cells) remain within brain tissue, thus failing to stimulate a systemic immune response. Immune privilege describes how the immune system responds to particulate antigens localized selectively within the brain parenchyma. We believe this immune privilege is caused by the absence of antigen presenting dendritic cells from the brain. We tested the prediction that expression of fms-like tyrosine kinase ligand 3 (Flt3L) in the brain will recruit dendritic cells and induce a systemic immune response against exogenous influenza hemagglutinin in BALB/c mice. Coexpression of Flt3L with HA in the brain parenchyma induced a robust systemic anti-HA immune response, and a small response against myelin basic protein and proteolipid protein epitopes. Depletion of CD4(+)CD25+ regulatory T cells (Tregs) enhanced both responses. To investigate the autoimmune impact of these immune responses, we characterized the neuropathological and behavioral consequences of intraparenchymal injections of Flt3L and HA in BALB/c and C57BL/6 mice. T cell infiltration in the forebrain was time and strain dependent, and increased in animals treated with Flt3L and depleted of Tregs; however, we failed to detect widespread defects in myelination throughout the forebrain or spinal cord. Results of behavioral tests were all normal. These results demonstrate that Flt3L overcomes the brain's immune privilege, and supports the clinical development of Flt3L as an adjuvant to stimulate clinically effective immune responses against brain neo-antigens, for example, those associated with brain tumors.


Asunto(s)
Encéfalo/inmunología , Sistema Inmunológico/inmunología , Tirosina Quinasa 3 Similar a fms/inmunología , Adyuvantes Inmunológicos , Animales , Antígenos/inmunología , Células Dendríticas/inmunología , Hemaglutininas/inmunología , Inmunidad , Ligandos , Ratones , Ratones Endogámicos BALB C , Prosencéfalo/inmunología , Médula Espinal/inmunología , Linfocitos T Reguladores/inmunología
10.
Ther Adv Neurol Disord ; 16: 17562864221150040, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36741352

RESUMEN

Background: Natalizumab is a highly effective monoclonal antibody for the treatment of multiple sclerosis (MS), which can diffuse in different anatomical compartments, including cerebrospinal fluid (CSF) and milk. Objectives: Starting from incidental detection of natalizumab in the CSF of MS patients, the objective of this study was to develope a flow-cytometry-based assay and apply it to quantify natalizumab in body fluids, including milk collected from nursing patients over 180 days and in patients with neutralizing antibodies against natalizumab. Methods: CSF, milk and sera samples from patients with multiple sclerosis were tested by flow-cytometry for binding to a VLA-4 expressing cell line or to a control cell line. A standard curve was prepared by incubating the same cells with natalizumab at 50 µg/ml and serially diluted to 0.005 ng/ml. Binding specificity was confirmed using an anti-natalizumab neutralizing antibody. Results: Our assay was sensitive enough to detect natalizumab in CSF, with a lower detection limit of 1.5 ng/ml. Neutralizing antibodies against natalizumab inhibited binding to the cell line. In breastmilk, the peak concentration was observed during the first 2 weeks after infusion and the average concentration over the observation time was 173.3 ng/ml, with a trend toward increased average milk concentration over subsequent administrations. Conclusion: Routine use of such an assay would enable a better understanding of the safety of therapeutic antibody administration during pregnancy and lactation.

11.
Mol Ther ; 19(10): 1793-801, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21505426

RESUMEN

Glioblastoma multiforme (GBM) is a primary brain tumor with a median survival of 14.6 months postdiagnosis. The infiltrative nature of GBM prevents complete resection and residual brain tumor cells give rise to recurrent GBM, a hallmark of this disease. Recurrent GBMs are known to harbor numerous mutations/gene rearrangements when compared to the primary tumor, which leads to the potential expression of novel proteins that could serve as tumor neoantigens. We have developed a combined immune-based gene therapeutic approach for GBM using adenoviral (Ads) mediated gene delivery of Herpes Simplex Virus Type 1-thymidine kinase (TK) into the tumor mass to induce tumor cells' death combined with an adenovirus expressing fms-like tyrosine kinase 3 ligand (Flt3L) to recruit dendritic cells (DCs) into the tumor microenvironment. This leads to the induction of specific anti-brain tumor immunity and immunological memory. In a model of GBM recurrence, we demonstrate that Flt3L/TK mediated immunological memory is capable of recognizing brain tumor neoantigens absent from the original treated tumor. These data demonstrate that the Flt3L/TK gene therapeutic approach can induce systemic immunological memory capable of recognizing a brain tumor neoantigen in a model of recurrent GBM.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/terapia , Timidina Quinasa/genética , Tirosina Quinasa 3 Similar a fms/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Linfocitos T/inmunología
12.
Mol Immunol ; 151: 11-18, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36063582

RESUMEN

Myasthenia gravis is a neuromuscular disease associated with antibodies against components of the neuromuscular junction, most often against the acetylcholine receptor (AChR). Although several mechanisms have been postulated to explain how these autoantibodies can lead to the pathology of the disease, convincing evidence suggests that destruction of the receptor-bearing postsynaptic membrane by complement membrane attack complex is of central importance. In this review, evidence for the importance of complement, and possible relationships between autoantigen, autoantibodies, complement activation, and the destruction of the membrane are discussed. More recent insights from the results of the complement-inhibiting therapeutic antibody eculizumab are also described, and the mechanisms connecting antibody binding to complement activation are considered from a structural viewpoint.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento , Miastenia Gravis , Autoanticuerpos , Autoantígenos , Proteínas del Sistema Complemento , Humanos , Receptores Colinérgicos/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-33589541

RESUMEN

OBJECTIVE: To identify an MS-specific immune cell population by deep immune phenotyping and relate it to soluble signaling molecules in CSF. METHODS: We analyzed surface expression of 22 markers in paired blood/CSF samples from 39 patients using mass cytometry (cytometry by time of flight). We also measured the concentrations of 296 signaling molecules in CSF using proximity extension assay. Results were analyzed using highly automated unsupervised algorithmic informatics. RESULTS: Mass cytometry objectively identified a B-cell population characterized by the expression of CD49d, CD69, CD27, CXCR3, and human leukocyte antigen (HLA)-DR as clearly associated with MS. Concentrations of the B cell-related factors, notably FCRL2, were increased in MS CSF, especially in early stages of the disease. The B-cell trophic factor B cell activating factor (BAFF) was decreased in MS. Proteins involved in neural plasticity were also reduced in MS. CONCLUSION: When analyzed without a priori assumptions, both the soluble and the cellular compartments of the CSF in MS were characterized by markers related to B cells, and the strongest candidate for an MS-specific cell type has a B-cell phenotype.


Asunto(s)
Factor Activador de Células B/líquido cefalorraquídeo , Linfocitos B/citología , Biomarcadores/líquido cefalorraquídeo , Esclerosis Múltiple/inmunología , Adulto , Linfocitos B/inmunología , Biomarcadores/análisis , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
14.
Front Immunol ; 10: 829, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31040853

RESUMEN

Determining antigen specificity is vital for understanding B cell biology and for producing human monoclonal antibodies. We describe here a powerful method for identifying B cells that recognize membrane antigens expressed on cells. The technique depends on two characteristics of the interaction between a B cell and an antigen-expressing cell: antigen-receptor-mediated extraction of antigen from the membrane of the target cell, and B cell activation. We developed the method using influenza hemagglutinin as a model viral membrane antigen, and tested it using acetylcholine receptor (AChR) as a model membrane autoantigen. The technique involves co-culturing B cells with adherent, bioorthogonally labeled cells expressing GFP-tagged antigen, and sorting GFP-capturing, newly activated B cells. Hemagglutinin-specific B cells isolated this way from vaccinated human donors expressed elevated CD20, CD27, CD71, and CD11c, and reduced CD21, and their secreted antibodies blocked hemagglutination and neutralized viral infection. Antibodies cloned from AChR-capturing B cells derived from patients with myasthenia gravis bound specifically to the receptor on cell membrane. The approach is sensitive enough to detect antigen-specific B cells at steady state, and can be adapted for any membrane antigen.


Asunto(s)
Antígenos de Superficie/inmunología , Linfocitos B/inmunología , Separación Celular/métodos , Adulto , Anciano , Animales , Antígenos de Superficie/aislamiento & purificación , Autoantígenos/inmunología , Autoantígenos/aislamiento & purificación , Subgrupos de Linfocitos B/inmunología , Línea Celular Tumoral , Células Clonales , Epítopos de Linfocito B/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunofenotipificación , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miastenia Gravis/inmunología , Receptores Colinérgicos/inmunología
15.
Eur J Neurosci ; 27(1): 183-90, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18184320

RESUMEN

Copulatory behaviors are generally dependent on testicular androgens in male vertebrates, being eliminated by castration and reinstated by testosterone administration. It is postulated that a critical factor in this hormonal gating is up-regulation of neuronal nitric oxide synthase (nNOS) in the preoptic area, and consequent enhanced nitric oxide synthesis in response to stimuli associated with a receptive female. Previous studies have suggested that nNOS protein is more abundant in behaviorally relevant preoptic regions of testosterone-exposed animals than in hormone-deprived controls. This study sought to elucidate the molecular events underlying this apparent up-regulation by examining preoptic nNOS mRNA abundance at several time points following testosterone administration in a castration and replacement paradigm. Castrated male whiptails (Cnemidophorus inornatus) were implanted with testosterone, and at four time points over the subsequent 18 days their sexual behavior was tested. A rostral periventricular area previously implicated in hormonal gating of male-typical copulatory behavior was then excised by laser microdissection, and nNOS transcript abundance was assessed by quantitative PCR. As neither this technique nor nNOS mRNA measurements have previously been performed in this area of the brain, expression was concomitantly assayed on adjacent sections by in situ hybridization or NADPH diaphorase histochemistry. Results are consistent with transcriptional up-regulation of nNOS by testosterone and a central role for the enzyme in mediating hormonal gating of copulatory behavior.


Asunto(s)
Andrógenos/farmacología , Copulación/efectos de los fármacos , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa/metabolismo , Área Preóptica/citología , Testosterona/farmacología , Análisis de Varianza , Animales , Conducta Animal , Recuento de Células , Masculino , NADPH Deshidrogenasa/metabolismo , Ratas , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
16.
Brain Res ; 1052(2): 236-9, 2005 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-16023092

RESUMEN

The synthesis of nitric oxide by the enzyme nitric oxide synthase (NOS) is involved in the androgen-dependent gating of male-typical copulatory behavior, both centrally, particularly in the preoptic area, and peripherally, notably through its role in penile erection. In the all-female whiptail lizard species Cnemidophorus uniparens, individuals display copulatory behaviors indistinguishable from males of similar species if gonadectomized and treated with testosterone. In this experiment, androgenized individuals were treated with a NOS inhibitor, which eliminated male-like behavior in half the individuals, suggesting that the central role of nitric oxide synthesis is conserved in this species. The deficit was principally in mounting, suggesting that sexual motivational systems were affected, rather than consummatory mechanisms.


Asunto(s)
Andrógenos/farmacología , Inhibidores Enzimáticos/farmacología , NG-Nitroarginina Metil Éster/farmacología , Conducta Sexual Animal/efectos de los fármacos , Testosterona/farmacología , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Interacciones Farmacológicas , Femenino , Lagartos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Ovariectomía/métodos , Tiempo de Reacción/efectos de los fármacos
17.
Neurol Neuroimmunol Neuroinflamm ; 2(5): e144, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26380353

RESUMEN

OBJECTIVE: To monitor the systemic immune responses of patients with multiple sclerosis (MS) under treatment with GNbAC1, a monoclonal antibody against the envelope protein of the MS- associated retrovirus, during a phase 2a trial. METHODS: We analyzed the composition of immune cell subsets and the activation level of monocytes by flow cytometry and the response against viral and vaccine antigens by ELISpot. RESULTS: None of the endpoints measured revealed any immunosuppressive effect of the therapeutic antibody. Activation of monocytes slightly decreased during treatment as predicted by the hypothesized mechanism of action of GNbAC1. CONCLUSION: These results support the conclusion that the antibody is safe for use in patients with MS. CLASSIFICATION OF EVIDENCE: This study provides Class III evidence that in patients with MS GNbAC1 does not significantly affect several biomarkers of systemic immune response.

18.
PLoS One ; 6(8): e23523, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21897844

RESUMEN

CD8(+) T cells infiltrate the brain during an anti-viral immune response. Within the brain CD8(+) T cells recognize cells expressing target antigens, become activated, and secrete IFNγ. However, there are no methods to recognize individual cells that respond to IFNγ. Using a model that studies the effects of the systemic anti-adenoviral immune response upon brain cells infected with an adenoviral vector in mice, we describe a method that identifies individual cells that respond to IFNγ. To identify individual mouse brain cells that respond to IFNγ we constructed a series of adenoviral vectors that contain a transcriptional response element that is selectively activated by IFNγ signaling, the gamma-activated site (GAS) promoter element; the GAS element drives expression of a transgene, Cre recombinase (Ad-GAS-Cre). Upon binding of IFNγ to its receptor, the intracellular signaling cascade activates the GAS promoter, which drives expression of the transgene Cre recombinase. We demonstrate that upon activation of a systemic immune response against adenovirus, CD8(+) T cells infiltrate the brain, interact with target cells, and cause an increase in the number of cells expressing Cre recombinase. This method can be used to identify, study, and eventually determine the long term fate of infected brain cells that are specifically targeted by IFNγ. The significance of this method is that it will allow to characterize the networks in the brain that respond to the specific secretion of IFNγ by anti-viral CD8(+) T cells that infiltrate the brain. This will allow novel insights into the cellular and molecular responses underlying brain immune responses.


Asunto(s)
Antivirales/metabolismo , Encéfalo/citología , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/metabolismo , Imagen Molecular/métodos , Transducción de Señal/inmunología , Adenoviridae/genética , Animales , Encéfalo/metabolismo , Encéfalo/virología , Femenino , Genes Reporteros/genética , Vectores Genéticos/genética , Herpesvirus Humano 1/genética , Integrasas/metabolismo , Interferón gamma/genética , Masculino , Ratones , Regiones Promotoras Genéticas/genética , Especificidad de la Especie , Timidina Quinasa/genética
19.
Curr Protoc Neurosci ; Chapter 4: Unit 4.24, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20066657

RESUMEN

Viral vector-mediated gene delivery is an attractive procedure for introducing genes into the brain, both for purposes of basic neuroscience research and to develop gene therapy for neurological diseases. Replication-defective adenoviruses possess many features which make them ideal vectors for this purpose-efficiently transducing terminally differentiated cells such as neurons and glial cells, resulting in high levels of transgene expression in vivo. Also, in the absence of anti-adenovirus immunity, these vectors can sustain very long-term transgene expression within the brain parenchyma. This unit provides protocols for the stereotactic injection of adenoviral vectors into the brain, followed by protocols to detect transgene expression or infiltrates of immune cells by immunocytochemistry or immunofluorescence. ELISPOT and neutralizing antibody assay methodologies are provided to quantitate the levels of cellular and humoral immune responses against adenoviruses. Quantitation of adenoviral vector genomes within the rat brain using qPCR is also described.


Asunto(s)
Adenoviridae/genética , Encéfalo/virología , Técnicas de Transferencia de Gen , Vectores Genéticos , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Encéfalo/patología , Técnica del Anticuerpo Fluorescente/métodos , Técnicas de Transferencia de Gen/efectos adversos , Vectores Genéticos/efectos adversos , Inmunohistoquímica/métodos , Neuroinmunomodulación , Reacción en Cadena de la Polimerasa/métodos , Ratas , Técnicas Estereotáxicas
20.
PLoS One ; 3(8): e2977, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18714338

RESUMEN

BACKGROUND: Astrocytes usually respond to trauma, stroke, or neurodegeneration by undergoing cellular hypertrophy, yet, their response to a specific immune attack by T cells is poorly understood. Effector T cells establish specific contacts with target cells, known as immunological synapses, during clearance of virally infected cells from the brain. Immunological synapses mediate intercellular communication between T cells and target cells, both in vitro and in vivo. How target virally infected astrocytes respond to the formation of immunological synapses established by effector T cells is unknown. FINDINGS: Herein we demonstrate that, as a consequence of T cell attack, infected astrocytes undergo dramatic morphological changes. From normally multipolar cells, they become unipolar, extending a major protrusion towards the immunological synapse formed by the effector T cells, and withdrawing most of their finer processes. Thus, target astrocytes become polarized towards the contacting T cells. The MTOC, the organizer of cell polarity, is localized to the base of the protrusion, and Golgi stacks are distributed throughout the protrusion, reaching distally towards the immunological synapse. Thus, rather than causing astrocyte hypertrophy, antiviral T cells cause a major structural reorganization of target virally infected astrocytes. CONCLUSIONS: Astrocyte polarization, as opposed to hypertrophy, in response to T cell attack may be due to T cells providing a very focused attack, and thus, astrocytes responding in a polarized manner. A similar polarization of Golgi stacks towards contacting T cells was also detected using an in vitro allogeneic model. Thus, different T cells are able to induce polarization of target astrocytes. Polarization of target astrocytes in response to immunological synapses may play an important role in regulating the outcome of the response of astrocytes to attacking effector T cells, whether during antiviral (e.g. infected during HIV, HTLV-1, HSV-1 or LCMV infection), anti-transplant, autoimmune, or anti-tumor immune responses in vivo and in vitro.


Asunto(s)
Astrocitos/fisiología , Encéfalo/fisiología , Sinapsis/fisiología , Linfocitos T/inmunología , Adenoviridae , Animales , Astrocitos/citología , Astrocitos/inmunología , Polaridad Celular/fisiología , Técnicas de Cocultivo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/fisiología , Cuerpo Estriado/virología , Masculino , Microscopía Confocal , Ratas , Ratas Sprague-Dawley , Bazo/citología , Bazo/inmunología , Bazo/fisiología , Bazo/virología , Timidina Quinasa/administración & dosificación , Timidina Quinasa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA