Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 140(2): 353-369, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27956400

RESUMEN

Ischaemic stroke induces endogenous repair processes that include proliferation and differentiation of neural stem cells and extensive rewiring of the remaining neural connections, yet about 50% of stroke survivors live with severe long-term disability. There is an unmet need for drug therapies to improve recovery by promoting brain plasticity in the subacute to chronic phase after ischaemic stroke. We previously showed that complement-derived peptide C3a regulates neural progenitor cell migration and differentiation in vitro and that C3a receptor signalling stimulates neurogenesis in unchallenged adult mice. To determine the role of C3a-C3a receptor signalling in ischaemia-induced neural plasticity, we subjected C3a receptor-deficient mice, GFAP-C3a transgenic mice expressing biologically active C3a in the central nervous system, and their respective wild-type controls to photothrombotic stroke. We found that C3a overexpression increased, whereas C3a receptor deficiency decreased post-stroke expression of GAP43 (P < 0.01), a marker of axonal sprouting and plasticity, in the peri-infarct cortex. To verify the translational potential of these findings, we used a pharmacological approach. Daily intranasal treatment of wild-type mice with C3a beginning 7 days after stroke induction robustly increased synaptic density (P < 0.01) and expression of GAP43 in peri-infarct cortex (P < 0.05). Importantly, the C3a treatment led to faster and more complete recovery of forepaw motor function (P < 0.05). We conclude that C3a-C3a receptor signalling stimulates post-ischaemic neural plasticity and intranasal treatment with C3a receptor agonists is an attractive approach to improve functional recovery after ischaemic brain injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Complemento C3a/uso terapéutico , Plasticidad Neuronal/efectos de los fármacos , Administración Intranasal , Animales , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/genética , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Lateralidad Funcional/efectos de los fármacos , Lateralidad Funcional/genética , Proteína GAP-43/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/genética , Desempeño Psicomotor/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Sinapsinas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
2.
J Immunol ; 192(10): 4469-74, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24795464

RESUMEN

ICAM-1 plays an important role in leukocyte trafficking, immunological synapse formation, and numerous cellular immune responses. Although considered a single glycoprotein, there are multiple membrane-bound and soluble ICAM-1 isoforms that arise from alternative splicing and proteolytic cleavage during inflammatory responses. The function and expression of these isoforms on various cell types are poorly understood. In the generation of ICAM-1-deficient mice, two isoform-deficient ICAM-1 mutants were inadvertently produced as a result of alternative splicing. These mice, along with true ICAM-1-deficient mice and newly generated ICAM-1-transgenic mice, have provided the opportunity to begin examining the role of ICAM-1 isoforms (singly or in combination) in various disease settings. In this review, we highlight the sharply contrasting disease phenotypes using ICAM-1 isoform mutant mice. These studies demonstrate that ICAM-1 immunobiology is highly complex but that individual isoforms, aside from the full-length molecule, make significant contributions to disease development and pathogenesis.


Asunto(s)
Sinapsis Inmunológicas/inmunología , Molécula 1 de Adhesión Intercelular/inmunología , Empalme Alternativo/genética , Empalme Alternativo/inmunología , Animales , Humanos , Sinapsis Inmunológicas/genética , Molécula 1 de Adhesión Intercelular/genética , Ratones , Ratones Transgénicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Proteolisis
3.
Proc Natl Acad Sci U S A ; 110(4): 1416-21, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23297203

RESUMEN

CD8 T-cell responses are critical for protection against intracellular pathogens and tumors. The induction and properties of these responses are governed by a series of integrated processes that rely heavily on cell-cell interactions. Intercellular adhesion molecule (ICAM)-1 functions to enhance the strength of antigenic stimulation, extend the duration of contact with antigen-presenting cells, and augment cytokine signals, which are all factors that influence peripheral CD8 T-cell differentiation. Although previous studies suggest that ICAM-1 is essential for establishing memory T-cell populations following peptide immunization, the roles of ICAM-1 in antiviral cellular immunity are less well understood. Here we show that, following a prototypic acute viral infection, the formation and maintenance of memory-phenotype CD127(hi), KLRG-1(lo) CD8 T cells does not require ICAM-1. Nevertheless, ICAM-1 expression on nonlymphocytes dictates the phenotypic and functional attributes of the antiviral CD8 T-cell populations that develop and promotes the gradual attrition of residual effector-like CD127(lo), KLRG-1(hi) CD8 T cells during the memory phase of the response. Although memory T cells do emerge and are maintained if ICAM-1 expression is abolished, the secondary proliferative capacity of these T cells is severely curtailed. Collectively, these studies reveal potential dual roles for ICAM-1 in both promoting the decay of effector responses and programming the sensitivity of memory CD8 T cells to secondary stimuli.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Memoria Inmunológica/fisiología , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Enfermedad Aguda , Animales , Molécula 1 de Adhesión Intercelular/genética , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Lectinas Tipo C , Activación de Linfocitos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Inmunológicos/metabolismo
4.
Eur J Immunol ; 44(4): 1194-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24435747

RESUMEN

Intercellular adhesion molecule-1 (ICAM-1) plays an important role in leukocyte trafficking, induction of cellular immune responses, and immunological synapse formation. As a member of the immunoglobulin superfamily of adhesion proteins, ICAM-1 is composed of repeating Ig-like domains, a transmembrane domain, and short cytoplasmic tail that participates in intracellular signaling events. At least seven ICAM-1 protein isoforms are generated by alternative splicing, however little is known regarding their immunobiology. We have previously shown using different lines of ICAM-1 mutant mice (Icam1(tm1Jcgr) and Icam1(tm1Bay) ) that expression of alternatively spliced ICAM-1 isoforms can significantly influence the disease course during the development of EAE. In this study, we show using a newly developed transgenic mouse (CD2-Icam1(D4del) /Icam1(null) ) that T-cell-specific expression of a single ICAM-1 isoform composed of Ig domains 1, 2, 3, and 5 can mediate the initiation and progression of EAE. Our results indicate that the ICAM-1 isoform lacking Ig domain 4 can drive pathogenesis in demyelinating disease and may be a novel therapeutic target for treating multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Molécula 1 de Adhesión Intercelular/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Empalme Alternativo , Animales , Sitios de Unión/genética , Sitios de Unión/inmunología , Western Blotting , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Citometría de Flujo , Células HEK293 , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Linfocitos T/metabolismo , Linfocitos T/trasplante
5.
J Biol Chem ; 288(16): 10962-6, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23493396

RESUMEN

Cerebral malaria (CM) is a severe clinical complication of Plasmodium falciparum malaria infection and is characterized by a high fatality rate and neurological damage. Sequestration of parasite-infected red blood cells in brain microvasculature utilizes host- and parasite-derived adhesion molecules and is an important factor in the development of CM. ICAM-1, an alternatively spliced adhesion molecule, is believed to be critical on endothelial cells for infected red blood cell sequestration in CM. Using ICAM-1 mutant mice, we found that the full-length ICAM-1 isoform is not required for development of murine experimental CM (ECM) and that ECM phenotype varies with the combination of ICAM-1 isoforms expressed. Furthermore, we observed development of ECM in transgenic mice expressing ICAM-1 only on leukocytes, indicating that endothelial cell expression of this adhesion molecule is not required for disease pathogenesis. We propose that ICAM-1-dependent cellular aggregation, independent of ICAM-1 expression on the cerebral microvasculature, contributes to ECM.


Asunto(s)
Encéfalo , Circulación Cerebrovascular , Molécula 1 de Adhesión Intercelular/biosíntesis , Malaria Cerebral/metabolismo , Microcirculación , Plasmodium falciparum/metabolismo , Empalme Alternativo/genética , Animales , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Encéfalo/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Molécula 1 de Adhesión Intercelular/genética , Malaria Cerebral/genética , Malaria Cerebral/patología , Malaria Cerebral/fisiopatología , Ratones , Ratones Transgénicos , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética
6.
FASEB J ; 27(9): 3797-804, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23737250

RESUMEN

Complement is an essential component of inflammation that plays a role in ischemic brain injury. Recent reports demonstrate novel functions of complement in normal and diseased CNS, such as regulation of neurogenesis and synapse elimination. Here, we examined the role of complement-derived peptide C3a in unilateral hypoxia-ischemia (HI), a model of neonatal HI encephalopathy. HI injury was induced at postnatal day 9 (P9), and loss of hippocampal tissue was determined on P31. We compared WT mice with transgenic mice expressing C3a under the control of glial fibrillary acidic protein promoter, which express biologically active C3a only in CNS and without the requirement of a priori complement activation. Further, we injected C3a peptide into the lateral cerebral ventricle of mice lacking the C3a receptor (C3aR) and WT mice and assessed HI-induced memory impairment 41 d later. We found that HI-induced tissue loss in C3a overexpressing mice was reduced by 50% compared with WT mice. C3a peptide injected 1 h after HI protected WT but not C3aR-deficient mice against HI-induced memory impairment. Thus, C3a acting through its canonical receptor ameliorates behavioral deficits after HI injury, and C3aR is a novel therapeutic target for the treatment of neonatal HI encephalopathy.


Asunto(s)
Hipoxia-Isquemia Encefálica/metabolismo , Receptores de Complemento/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/metabolismo , Complemento C3a/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Inmunohistoquímica , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/genética , Receptores de Complemento/deficiencia , Receptores de Complemento/genética
7.
Epilepsia ; 55(12): e139-42, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25385326

RESUMEN

Studies have demonstrated that the membrane attack complex (MAC) of complement can evoke seizures when injected directly into rodent brain. In the course of studies that examine the role of complement in the development of experimental cerebral malaria (ECM), we observed fewer seizures in mice deficient in C5, a component required for MAC formation. To determine if the MAC contributed to the tonic-clonic seizures characteristic of ECM, we performed long-term video-electroencephalography (EEG) on C5(-/-) mice with Plasmodium berghei ANKA-induced cerebral malaria and observed significantly reduced spike and seizure frequency compared to wild-type mice. Our data suggest a role for the MAC in malaria-induced seizures and that inhibition of the terminal complement pathway may reduce seizures and seizure-related neurocognitive deficits.


Asunto(s)
Complemento C5/deficiencia , Malaria Cerebral/complicaciones , Convulsiones , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Malaria Cerebral/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium berghei/patogenicidad , Convulsiones/etiología , Convulsiones/genética , Convulsiones/prevención & control
8.
J Biol Chem ; 287(29): 24734-8, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22689574

RESUMEN

Cerebral malaria (CM) is the most severe manifestation of clinical malaria syndromes and has a high fatality rate especially in the developing world. Recent studies demonstrated that C5(-/-) mice are resistant to experimental CM (ECM) and that protection was due to the inability to form the membrane attack complex. Unexpectedly, we observed that C4(-/-) and factor B(-/-) mice were fully susceptible to disease, indicating that activation of the classical or alternative pathways is not required for ECM. C3(-/-) mice were also susceptible to ECM, indicating that the canonical C5 convertases are not required for ECM development and progression. Abrogation of ECM by treatment with anti-C9 antibody and detection of C5a in serum of C3(-/-) mice confirmed that C5 activation occurs in ECM independent of C5 convertases. Our data indicate that activation of C5 in ECM likely occurs via coagulation enzymes of the extrinsic protease pathway.


Asunto(s)
Convertasas de Complemento C3-C5/metabolismo , Malaria Cerebral/inmunología , Malaria Cerebral/metabolismo , Animales , Activación de Complemento/genética , Activación de Complemento/fisiología , Complemento C3/genética , Complemento C3/metabolismo , Convertasas de Complemento C3-C5/genética , Complemento C4/genética , Complemento C4/metabolismo , Factor B del Complemento/genética , Factor B del Complemento/metabolismo , Malaria Cerebral/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Plasmodium berghei/inmunología , Plasmodium berghei/patogenicidad
9.
J Immunol ; 186(12): 6657-60, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21572031

RESUMEN

Cerebral malaria is the most severe complication of Plasmodium falciparum infection and accounts for a large number of malaria fatalities worldwide. Recent studies demonstrated that C5(-/-) mice are resistant to experimental cerebral malaria (ECM) and suggested that protection was due to loss of C5a-induced inflammation. Surprisingly, we observed that C5aR(-/-) mice were fully susceptible to disease, indicating that C5a is not required for ECM. C3aR(-/-) and C3aR(-/-) × C5aR(-/-) mice were equally susceptible to ECM as were wild-type mice, indicating that neither complement anaphylatoxin receptor is critical for ECM development. In contrast, C9 deposition in the brains of mice with ECM suggested an important role for the terminal complement pathway. Treatment with anti-C9 Ab significantly increased survival time and reduced mortality in ECM. Our data indicate that protection from ECM in C5(-/-) mice is mediated through inhibition of membrane attack complex formation and not through C5a-induced inflammation.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Malaria Cerebral/etiología , Animales , Encéfalo/inmunología , Complemento C5a/fisiología , Complemento C9/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Susceptibilidad a Enfermedades , Inflamación , Malaria Cerebral/inmunología , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a , Receptores de Complemento , Tasa de Supervivencia
10.
Biochem J ; 446(3): 499-508, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22720637

RESUMEN

Storage of erythrocytes in blood banks is associated with biochemical and morphological changes to RBCs (red blood cells). It has been suggested that these changes have potential negative clinical effects characterized by inflammation and microcirculatory dysfunction which add to other transfusion-related toxicities. However, the mechanisms linking RBC storage and toxicity remain unclear. In the present study we tested the hypothesis that storage of leucodepleted RBCs results in cells that inhibit NO (nitric oxide) signalling more so than younger cells. Using competition kinetic analyses and protocols that minimized contributions from haemolysis or microparticles, our data indicate that the consumption rates of NO increased ~40-fold and NO-dependent vasodilation was inhibited 2-4-fold comparing 42-day-old with 0-day-old RBCs. These results are probably due to the formation of smaller RBCs with increased surface area: volume as a consequence of membrane loss during storage. The potential for older RBCs to affect NO formation via deoxygenated RBC-mediated nitrite reduction was also tested. RBC storage did not affect deoxygenated RBC-dependent stimulation of nitrite-induced vasodilation. However, stored RBCs did increase the rates of nitrite oxidation to nitrate in vitro. Significant loss of whole-blood nitrite was also observed in stable trauma patients after transfusion with 1 RBC unit, with the decrease in nitrite occurring after transfusion with RBCs stored for >25 days, but not with younger RBCs. Collectively, these data suggest that increased rates of reactions between intact RBCs and NO and nitrite may contribute to mechanisms that lead to storage-lesion-related transfusion risk.


Asunto(s)
Eritrocitos/citología , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Animales , Conservación de la Sangre/métodos , Adhesión Celular , Deformación Eritrocítica , Eritrocitos/fisiología , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Vasodilatación
11.
Front Immunol ; 13: 816640, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35178049

RESUMEN

Periodontitis is a dysbiotic infectious disease that leads to the destruction of tooth supporting tissues. There is increasing evidence that periodontitis may affect the development and severity of Alzheimer's disease (AD). However, the mechanism(s) by which periodontal infection impacts the neurodegenerative process in AD remains unclear. In the present study, using an amyloid precursor protein (APP) knock-in (App KI) AD mouse model, we showed that oral infection with Porphyromonas gingivalis (Pg), a keystone pathogen of periodontitis, worsened behavioral and cognitive impairment and accelerated amyloid beta (Aß) accumulation in AD mice, thus unquestionably and significantly aggravating AD. We also provide new evidence that the neuroinflammatory status established by AD, is greatly complicated by periodontal infection and the consequential entry of Pg into the brain via Aß-primed microglial activation, and that Pg-induced brain overactivation of complement C1q is critical for periodontitis-associated acceleration of AD progression by amplifying microglial activation, neuroinflammation, and tagging synapses for microglial engulfment. Our study renders support for the importance of periodontal infection in the innate immune regulation of AD and the possibility of targeting microbial etiology and periodontal treatment to ameliorate the clinical manifestation of AD and lower AD prevalence.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Complemento C1q/metabolismo , Microglía/metabolismo , Periodontitis/metabolismo , Periodontitis/microbiología , Sinapsis/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Porphyromonas gingivalis
12.
Transfusion ; 51(4): 867-73, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21496048

RESUMEN

The specific negative clinical manifestations associated with the transfusion of stored red blood cells (RBCs) and the corresponding mechanisms responsible for such phenomena remain poorly defined. Our recent studies document that leukoreduced older RBC units potentiate transfusion-related toxicity in trauma patients. It is our hypothesis that the transfusion of relatively older blood impedes microvascular perfusion. The central mechanisms proposed to mediate this microcirculatory alteration include: 1) the loss of RBC-dependent control of nitric oxide-mediated homeostasis concerning vasodilation and 2) immune cell and complement activation. In this review, we outline the background for our hypothesis and detail our current investigations toward the understanding of this pathophysiology.


Asunto(s)
Transfusión de Eritrocitos/efectos adversos , Eritrocitos/citología , Eritrocitos/metabolismo , Heridas y Lesiones/terapia , Conservación de la Sangre/efectos adversos , Humanos , Heridas y Lesiones/complicaciones
13.
J Immunol ; 182(5): 2628-40, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19234157

RESUMEN

In the CNS, the transcription factor NF-kappaB is a key regulator of inflammation and secondary injury processes. Following trauma or disease, the expression of NF-kappaB-dependent genes is activated, leading to both protective and detrimental effects. In this study, we show that transgenic inactivation of astroglial NF-kappaB (glial fibrillary acidic protein-IkappaB alpha-dominant-negative mice) resulted in reduced disease severity and improved functional recovery following experimental autoimmune encephalomyelitis. At the chronic stage of the disease, transgenic mice exhibited an overall higher presence of leukocytes in spinal cord and brain, and a markedly higher percentage of CD8(+)CD122(+) T regulatory cells compared with wild type, which correlated with the timing of clinical recovery. We also observed that expression of proinflammatory genes in both spinal cord and cerebellum was delayed and reduced, whereas the loss of neuronal-specific molecules essential for synaptic transmission was limited compared with wild-type mice. Furthermore, death of retinal ganglion cells in affected retinas was almost abolished, suggesting the activation of neuroprotective mechanisms. Our data indicate that inhibiting NF-kappaB in astrocytes results in neuroprotective effects following experimental autoimmune encephalomyelitis, directly implicating astrocytes in the pathophysiology of this disease.


Asunto(s)
Astrocitos/inmunología , Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Mediadores de Inflamación/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Enfermedad Crónica , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/fisiopatología , Femenino , Perfilación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/deficiencia , Proteína Ácida Fibrilar de la Glía/genética , Proteínas I-kappa B/deficiencia , Proteínas I-kappa B/genética , Mediadores de Inflamación/fisiología , Masculino , Ratones , Ratones Transgénicos , Inhibidor NF-kappaB alfa , FN-kappa B/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Índice de Severidad de la Enfermedad
14.
Eur J Immunol ; 39(6): 1516-26, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19384874

RESUMEN

We have previously shown that gammadelta T cells traffic to the CNS during EAE with concurrently increased expression of beta(2)-integrins and production of IFN-gamma and TNF-alpha. To extend these studies, we transferred bioluminescent gammadelta T cells to WT mice and followed their movement through the acute stages of disease. We found that gammadelta T cells rapidly migrated to the site of myelin oligodendrocyte glycoprotein peptide injection and underwent massive expansion. Within 6 days after EAE induction, bioluminescent gammadelta T cells were found in the spinal cord and brain, peaking in number between days 10 and 12 and then rapidly declining by day 15. Reconstitution of gammadelta T cell(-/-) mice with gammadelta T cells derived from beta(2)-integrin-deficient mice (CD11a, -b or -c) demonstrated that gammadelta T-cell trafficking to the CNS during EAE is independent of this family of adhesion molecules. We also examined the role of gammadelta T-cell-produced IFN-gamma and TNF-alpha in EAE and found that production of both cytokines by gammadelta T cells was required for full development of EAE. These results indicate that gammadelta T cells are critical for the development of EAE and suggest a therapeutic target in demyelinating disease.


Asunto(s)
Movimiento Celular/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/fisiopatología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Encéfalo/inmunología , Encéfalo/patología , Antígenos CD11/genética , Antígenos CD18/metabolismo , Movimiento Celular/genética , Citocinas/genética , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/diagnóstico , Glicoproteínas/administración & dosificación , Glicoproteínas/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Antígeno-1 Asociado a Función de Linfocito/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Médula Espinal/inmunología , Médula Espinal/patología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/trasplante , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Neurosci Res ; 88(1): 1-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19610090

RESUMEN

gammadelta T cells represent a small subpopulation of T cells expressing a restricted repertoire of T-cell receptors and, unlike alphabeta T cells, function more as cells of the innate immune system. These cells are found in skin and mucosal sites as well as secondary lymphoid tissues and frequently act as first line of defense sentinels. gammadelta T cells have been implicated in the pathogenesis of demyelinating disease, although little was known regarding their trafficking and effector functions. In this Mini-Review, we highlight recent studies demonstrating that gammadelta T cells migrate rapidly to the CNS during experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis. gammadelta T-cell trafficking to the CNS is independent of beta(2)-integrins and occurs well before onset of clinical signs of disease, peaking early during the acute phase of disease. gammadelta T-cell-mediated production of inflammatory cytokines, including interferon-gamma and tumor necrosis factor-alpha, appears critical for EAE development, suggesting that these cells may set the stage for activation of other subsets of infiltrating effector cells. These data suggest that gammadelta T cells or subsets of gammadelta T cells may represent a new therapeutic target in demeylinating disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Citocinas/inmunología , Esclerosis Múltiple/inmunología
16.
Mol Immunol ; 46(5): 1007-10, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18952296

RESUMEN

Purification of lymphocytes, particularly T cells, is commonly performed using nylon wool. This enrichment method selectively retains B cells and some myeloid cells allowing a significantly more pure T cell population to flow through a nylon wool column. T cells purified in this fashion are assumed to be unaltered and functionally naïve, however some studies have suggested aberrant in vitro T cell responses after nylon wool treatment. We found that nylon wool purification significantly altered T cell proliferation, expression of activation markers and production of cytokines. Our results suggest that nylon wool treatment modifies T cell activation responses and that caution should be used when choosing this purification method.


Asunto(s)
Proliferación Celular , Activación de Linfocitos , Nylons , Linfocitos T/citología , Animales , Antígenos de Diferenciación/inmunología , Separación Celular , Ratones , Linfocitos T/inmunología
17.
Front Immunol ; 11: 585108, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240274

RESUMEN

The soluble membrane attack complex (sMAC, a.k.a., sC5b-9 or TCC) is generated on activation of complement and contains the complement proteins C5b, C6, C7, C8, C9 together with the regulatory proteins clusterin and/or vitronectin. sMAC is a member of the MACPF/cholesterol-dependent-cytolysin superfamily of pore-forming molecules that insert into lipid bilayers and disrupt cellular integrity and function. sMAC is a unique complement activation macromolecule as it is comprised of several different subunits. To date no complement-mediated function has been identified for sMAC. sMAC is present in blood and other body fluids under homeostatic conditions and there is abundant evidence documenting changes in sMAC levels during infection, autoimmune disease and trauma. Despite decades of scientific interest in sMAC, the mechanisms regulating its formation in healthy individuals and its biological functions in both health and disease remain poorly understood. Here, we review the structural differences between sMAC and its membrane counterpart, MAC, and examine sMAC immunobiology with respect to its presence in body fluids in health and disease. Finally, we discuss the diagnostic potential of sMAC for diagnostic and prognostic applications and potential utility as a companion diagnostic.


Asunto(s)
Activación de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Animales , Humanos
18.
Immunobiology ; 225(4): 151983, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32747015

RESUMEN

Dysregulated activation of inflammatory signaling by the immature neonatal immune system could lead to the development of many pediatric diseases including necrotizing enterocolitis (NEC). While the mechanism(s) of pathogenesis is unknown, NEC is believed to have multifactorial causes. Microbial dysbiosis and intestinal immaturity have been implicated as potential triggers for this disease. We hypothesized that psychological stress during pregnancy negatively impacts the development of intestinal tissues in offspring and contributes to development of NEC. Consistent with this hypothesis, we previously observed shorter villi and a decrease in total surface area in the small intestine of pups derived from mice that were chronically stressed during gestation. In this study, we performed RNASeq analysis to determine the gene expression changes in the offspring gut following prenatal stress in pregnant mice and identified several differentially expressed genes (DEGs) and biological pathways. Notably, C3 was upregulated in the small intestine and contributed to a higher tissue injury score in a mesenteric ischemia model compared to unstressed controls. We discuss the potential implications of these stress-induced genes expression changes and their contribution to development of intestinal inflammation.


Asunto(s)
Complemento C3/genética , Regulación de la Expresión Génica , Intestino Delgado/metabolismo , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/genética , Estrés Fisiológico , Animales , Complemento C3/inmunología , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/etiología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Intestino Delgado/inmunología , Isquemia Mesentérica/etiología , Ratones , Embarazo , Análisis de Secuencia de ARN
19.
J Neuroimmunol ; 206(1-2): 22-7, 2009 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-19010554

RESUMEN

LFA-1 (CD11a/CD18) is a member of the beta(2)-integrin family of adhesion molecules important in leukocyte trafficking and activation. Although LFA-1 is thought to contribute to the development of experimental autoimmune encephalomyelitis (EAE) primarily through its functions on effector T cells, its importance on other leukocyte populations remains unexplored. To address this question, we performed both adoptive transfer EAE experiments involving CD11a(-/-) mice and trafficking studies using bioluminescent T cells expressing luciferase under the control of a CD2 promoter (T-lux cells). Transfer of encephalitogenic CD11a(-/-) T cells to wild type mice resulted in a significant reduction in overall EAE severity compared to control transfers. We also observed, using in vivo imaging techniques, that CD11a(-/-) T-lux cells readily infiltrated lymph nodes and the CNS of wild type recipients with kinetics comparable to CD11a(+/+) transfers, although their overall numbers in these organs were reduced. Surprisingly, transfer of encephalitogenic wild type T cells to CD11a(-/-) mice induced a severe and sometimes fatal EAE disease course, associated with massive T cell infiltration and proliferation in the CNS. These data indicate that LFA-1 expression on leukocytes in recipient mice plays an important immunomodulatory role in EAE. Thus, LFA-1 acts as a key regulatory adhesion molecule during the development of EAE, serving both pro- and anti-inflammatory roles in disease pathogenesis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Antígeno-1 Asociado a Función de Linfocito/fisiología , Análisis de Varianza , Animales , Antígeno CD11a/genética , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/fisiopatología , Glicoproteínas/efectos adversos , Luciferasas/genética , Activación de Linfocitos/inmunología , Antígeno-1 Asociado a Función de Linfocito/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/efectos adversos , Estadísticas no Paramétricas , Linfocitos T/fisiología , Factores de Tiempo
20.
J Neuroimmunol ; 207(1-2): 18-23, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19135725

RESUMEN

Lysophosphatidylcholine (LPC) is a chemotactic lysolipid produced during inflammation by the hydrolytic action of phospholipase A(2) enzymes. LPC stimulates chemotaxis of T cells in vitro through activation of the G protein-coupled receptor, G2A. This has led to the proposition that G2A contributes to the recruitment of T cells to sites of inflammation and thus promotes chronic inflammatory autoimmune diseases associated with the generation and subsequent tissue infiltration of auto-antigen-specific effector T cells. However, one study suggests that G2A may negatively regulate T cell proliferative responses to antigen receptor engagement and thereby attenuates autoimmunity by reducing the generation of autoreactive T cells. To address the relative contribution of these G2A-mediated effects to the pathophysiology of T cell-mediated autoimmune disease, we examined the impact of G2A inactivation on the onset and severity of murine experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). Wild type (G2A(+/+)) and G2A-deficient (G2A(-/-)) C57BL/6J mice exhibited a similar incidence and onset of disease following immunization with MOG(35-55) peptide. Disease severity was only moderately reduced in G2A(-/-) mice. Similar numbers of MOG(35-55) specific T cells were generated in secondary lymphoid organs of MOG(35-55)-immunized G2A(+/+) and G2A(-/-) mice. Comparable numbers of T cells were detected in spinal cords of G2A(+/+) and G2A(-/-) mice. We conclude that the proposed anti-proliferative and chemotactic functions of G2A are not manifested in vivo and therefore therapeutic targeting of G2A is unlikely to be beneficial in the treatment of MS.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Receptores Acoplados a Proteínas G/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inducido químicamente , Citometría de Flujo/métodos , Eliminación de Gen , Glicoproteínas/efectos adversos , Interferón gamma/metabolismo , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/efectos adversos , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Bazo/citología , Bazo/inmunología , Bazo/patología , Linfocitos T/inmunología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA