13 research outputs found

    Are microglial cells the regulators of lymphocyte responses in the CNS?

    Get PDF
    The infiltration of immune cells in the central nervous system is a common hallmark in different neuroinflammatory conditions. Accumulating evidence indicates that resident glial cells can establish a cross-talk with infiltrated immune cells, including T-cells, regulating their recruitment, activation and function within the CNS. Although the healthy CNS has been thought to be devoid of professional dendritic cells (DCs), numerous studies have reported the presence of a population of DCs in specific locations such as the meninges, choroid plexuses and the perivascular space. Moreover, the infiltration of DC precursors during neuroinflammatory situations has been proposed, suggesting a putative role of these cells in the regulation of lymphocyte activity within the CNS. On the other hand, under specific circumstances, microglial cells are able to acquire a phenotype of DC expressing a wide range of molecules that equip these cells with all the necessary machinery for communication with T-cells. In this review, we summarize the current knowledge on the expression of molecules involved in the cross-talk with T-cells in both microglial cells and DCs and discuss the potential contribution of each of these cell populations on the control of lymphocyte function within the CNS

    Chronic IL-10 overproduction disrupts microglia-neuron dialogue similar to aging, resulting in impaired hippocampal neurogenesis and spatial memory

    Get PDF
    Altres ajuts: Acord transformatiu CRUE-CSICThis work was supported by the Spanish Ministry of Economy and Business (BFU2014-55459 and BFU2017-87843-R).The subgranular zone of the dentate gyrus is an adult neurogenic niche where new neurons are continuously generated. A dramatic hippocampal neurogenesis decline occurs with increasing age, contributing to cognitive deficits. The process of neurogenesis is intimately regulated by the microenvironment, with inflammation being considered a strong negative factor for this process. Thus, we hypothesize that the reduction of new neurons in the aged brain could be attributed to the age-related microenvironmental changes towards a pro-inflammatory status. In this work, we evaluated whether an anti-inflammatory microenvironment could counteract the negative effect of age on promoting new hippocampal neurons. Surprisingly, our results show that transgenic animals chronically overexpressing IL-10 by astrocytes present a decreased hippocampal neurogenesis in adulthood. This results from an impairment in the survival of neural newborn cells without differences in cell proliferation. In parallel, hippocampal-dependent spatial learning and memory processes were affected by IL-10 overproduction as assessed by the Morris water maze test. Microglial cells, which are key players in the neurogenesis process, presented a different phenotype in transgenic animals characterized by high activation together with alterations in receptors involved in neuronal communication, such as CD200R and CX3CR1. Interestingly, the changes described in adult transgenic animals were similar to those observed by the effect of normal aging. Thus, our data suggest that chronic IL-10 overproduction mimics the physiological age-related disruption of the microglia-neuron dialogue, resulting in hippocampal neurogenesis decrease and spatial memory impairment

    Differential Roles of TREM2+ Microglia in Anterograde and Retrograde Axonal Injury Models

    Get PDF
    Microglia are the main immune cells of the central nervous system (CNS), and they are devoted to the active surveillance of the CNS during homeostasis and disease. In the last years, the microglial receptor Triggering Receptor Expressed on Myeloid cells-2 (TREM2) has been defined to mediate several microglial functions, including phagocytosis, survival, proliferation, and migration, and to be a key regulator of a new common microglial signature induced under neurodegenerative conditions and aging, also known as disease-associated microglia (DAM). Although microglial TREM2 has been mainly studied in chronic neurodegenerative diseases, few studies address its regulation and functions in acute inflammatory injuries. In this context, the present work aims to study the regulation of TREM2 and its functions after reparative axonal injuries, using two-well established animal models of anterograde and retrograde neuronal degeneration: the perforant pathway transection (PPT) and the facial nerve axotomy (FNA). Our results indicate the appearance of a subpopulation of microglia expressing TREM2 after both anterograde and retrograde axonal injury. TREM2+ microglia were not directly related to proliferation, instead, they were associated with specific recognition and/or phagocytosis of myelin and degenerating neurons, as assessed by immunohistochemistry and flow cytometry. Characterization of TREM2+ microglia showed expression of CD16/32, CD68, and occasional Galectin-3. However, specific singularities within each model were observed in P2RY12 expression, which was only downregulated after PPT, and in ApoE, where de novo expression was detected only in TREM2+ microglia after FNA. Finally, we report that the pro-inflammatory or anti-inflammatory cytokine microenvironment, which may affect phagocytosis, did not directly modify the induction of TREM2+ subpopulation in any injury model, although it changed TREM2 levels due to modification of the microglial activation pattern. In conclusion, we describe a unique TREM2+ microglial subpopulation induced after axonal injury, which is directly associated with phagocytosis of specific cell remnants and show different phenotypes, depending on the microglial activation status and the degree of tissue injury

    From Mouse To Human : Comparative Analysis Between Grey And White Matter By Synchrotron-Fourier Transformed Infrared Microspectroscopy

    Get PDF
    Fourier Transform Infrared microspectroscopy (渭FTIR) is a very useful method to analyze the biochemical properties of biological samples in situ. Many diseases affecting the central nervous system (CNS) have been studied using this method, to elucidate alterations in lipid oxidation or protein aggregation, among others. In this work, we describe in detail the characteristics between grey matter (GM) and white matter (WM) areas of the human brain by 渭FTIR, and we compare them with the mouse brain (strain C57BL/6), the most used animal model in neurological disorders. Our results show a clear different infrared profile between brain areas in the lipid region of both species. After applying a second derivative in the data, we established a 1.5 threshold value for the lipid/protein ratio to discriminate between GM and WM areas in non-pathological conditions. Furthermore, we demonstrated intrinsic differences of lipids and proteins by cerebral area. Lipids from GM present higher C=CH, C=O and CH3 functional groups compared to WM in humans and mice. Regarding proteins, GM present lower Amide II amounts and higher intramolecular 尾-sheet structure amounts with respect to WM in both species. However, the presence of intermolecular 尾-sheet structures, which is related to 尾-aggregation, was only observed in the GM of some human individuals. The present study defines the relevant biochemical properties of non-pathological human and mouse brains by 渭FTIR as a benchmark for future studies involving CNS pathological samples

    TRPV2: A Key Player in Myelination Disorders of the Central Nervous System

    Get PDF
    Transient potential receptor vanilloid 2 (TRPV2) is widely expressed through the nervous system and specifically found in neuronal subpopulations and some glial cells. TRPV2 is known to be sensitized by methionine oxidation, which results from inflammation. Here we aim to characterize the expression and regulation of TRPV2 in myelination pathologies, such as hypomyelination and demyelination. We validated the interaction between TRPV2 and its putative interactor Opalin, an oligodendrocyte marker, in mixed glial cultures under pro- and anti-inflammatory conditions. Then, we characterized TRPV2 time-course expression in experimental animal models of hypomyelination (jimpy mice) and de-/remyelination (cuprizone intoxication and experimental autoimmune encephalomyelitis (EAE)). TRPV2 showed upregulation associated with remyelination, inflammation in cuprizone and EAE models, and downregulation in hypomyelinated jimpy mice. TRPV2 expression was altered in human samples of multiple sclerosis (MS) patients. Additionally, we analyzed the expression of methionine sulfoxide reductase A (MSRA), an enzyme that reduces oxidated methionines in TRPV2, which we found increased in inflammatory conditions. These results suggest that TRPV2 may be a key player in myelination in accordance with the recapitulation hypothesis, and that it may become an interesting clinical target in the treatment of demyelination disorders

    TRPV2 : a Key Player in Myelination Disorders of the Central Nervous System

    Get PDF
    Transient potential receptor vanilloid 2 (TRPV2) is widely expressed through the nervous system and specifically found in neuronal subpopulations and some glial cells. TRPV2 is known to be sensitized by methionine oxidation, which results from inflammation. Here we aim to characterize the expression and regulation of TRPV2 in myelination pathologies, such as hypomyelination and demyelination. We validated the interaction between TRPV2 and its putative interactor Opalin, an oligodendrocyte marker, in mixed glial cultures under pro- and anti-inflammatory conditions. Then, we characterized TRPV2 time-course expression in experimental animal models of hypomyelination (jimpy mice) and de-/remyelination (cuprizone intoxication and experimental autoimmune encephalomyelitis (EAE)). TRPV2 showed upregulation associated with remyelination, inflammation in cuprizone and EAE models, and downregulation in hypomyelinated jimpy mice. TRPV2 expression was altered in human samples of multiple sclerosis (MS) patients. Additionally, we analyzed the expression of methionine sulfoxide reductase A (MSRA), an enzyme that reduces oxidated methionines in TRPV2, which we found increased in inflammatory conditions. These results suggest that TRPV2 may be a key player in myelination in accordance with the recapitulation hypothesis, and that it may become an interesting clinical target in the treatment of demyelination disorders

    Papel de la microgl铆a en la regulaci贸n de la respuesta inmunitaria adquirida

    No full text
    Numerosos estudios han demostrado a lo largo de los a帽os, el papel fundamental que juegan las c茅lulas de microgl铆a en el funcionamiento del SNC. No s贸lo en condiciones normales, donde controlan la correcta home贸stasis del tejido, sino tambi茅n en todas aquellas situaciones que, como consecuencia de alteraciones y procesos patol贸gicos diversos, conllevan a una p茅rdida de esta home贸stasis. En respuesta a todas estas situaciones, las c茅lulas de microgl铆a son capaces de detectar r谩pidamente el da帽o y actuar de una manera espec铆fica en funci贸n del tipo de perturbaci贸n que se produzca en su entorno. Esta respuesta microglial ha sido ampliamente estudiada en da帽os agudos, en los que la microgl铆a act煤a como parte del sistema inmune innato, sin embargo los procesos que subyacen a la reactividad microglial ante una situaci贸n de inmunidad adquirida, as铆 como la comunicaci贸n que se establece entre estas c茅lulas microgliales y las c茅lulas inmunes perif茅ricas infiltradas permanecen en muchos aspectos sin esclarecer. En el presente trabajo hemos caracterizado el patr贸n de reactividad microglial y su relaci贸n con las diferentes poblaciones de linfocitos infiltrados a lo largo de las diferentes fases de la evoluci贸n que acontecen en un modelo agudo de encefalopat铆a autoinmune experimental (EAE). Nuestros estudios demuestran que las c茅lulas de microgl铆a se activan en respuesta a la inducci贸n de la EAE y presentan un patr贸n de activaci贸n espec铆fico en cada una de las fases. Durante la fase de inducci贸n y pico del proceso patol贸gico, en estrecha relaci贸n con el aumento de la sintomatolog铆a cl铆nica que se manifiesta por un progresivo deterioro de las funciones motoras, estas c茅lulas microgliales experimentan cambios morfol贸gicos y en su distribuci贸n acumul谩ndose alrededor de los vasos sangu铆neos. Adem谩s de microgl铆a reactiva y posiblemente de macr贸fagos de origen sangu铆neo, se observa tambi茅n un gran n煤mero de linfocitos infiltrados, mayoritariamente del tipo T-cooperador y subtipo Th1 (pro-inflamatorio), si bien tambi茅n se observan linfocitos T-citot贸xicos y T-γδ. En estas fases, la activaci贸n microglial se caracteriza a nivel fenot铆pico por el aumento en la expresi贸n de mol茅culas del complejo mayor de histocompatibilidad clase I y clase II (MHC-clase I y MHC-clase II) sin expresi贸n concomitante de mol茅culas co-estimuladoras B7.1 o B7.2, es decir las c茅lulas de microgl铆a activadas presentan en estas circunstancias un fenotipo caracter铆stico de c茅lulas dendr铆ticas inmaduras; hecho que hemos corroborado con la demostraci贸n de la expresi贸n del marcador CD1. En este contexto, la se帽al que inducen estas c茅lulas de microgl铆a a los linfocitos infiltrados, podr铆a estar implicada en la modulaci贸n del proceso inflamatorio induciendo la apoptosis o anergia linfocitaria. Durante la fase de recuperaci贸n, a pesar de que los animales experimentan una progresiva mejora sintomatol贸gica, las c茅lulas de microgl铆a siguen mostrando una morfolog铆a, distribuci贸n y patr贸n de expresi贸n de mol茅culas caracter铆sticos de c茅lulas reactivas. Si bien, en general, las c茅lulas de microgl铆a siguen mostrando el mismo patr贸n fenot铆pico (CD1+, MHC-I+, MHC-II+, B7.1- y B7.2-), aquellas localizadas en el entorno de algunos vasos sangu铆neos expresan diferencialmente la mol茅cula B7.2. Durante esta fase, adem谩s, el n煤mero total de linfocitos T-cooperadores, T-citot贸xicos y T-γδ se mantiene muy elevado con valores similares a los observados en las fases anteriores. Es interesante se帽alar que ya no encontramos linfocitos Th1 y la poblaci贸n de T-cooperadores est谩 constituida por los subtipos Th17 y T-regs. Los linfocitos que se acumulan en las inmediaciones de los vasos sangu铆neos expresan CTLA-4, uno de los co-receptores de B7.2. En este nuevo contexto, la interacci贸n de las c茅lulas de microgl铆a B7.2+ con estos linfocitos CTLA-4+ podr铆a ser la responsable de la resoluci贸n de la respuesta immunitaria y la inducci贸n de la tolerancia. En su conjunto los resultados obtenidos evidencian que la microgl铆a juega un papel clave en la evoluci贸n de la respuesta inmune adquirida modulando la activaci贸n e inactivaci贸n de las diferentes poblaciones linfocitarias implicadas tanto en la inducci贸n del proceso inmune/inflamatorio como en su posterior resoluci贸n. La realizaci贸n de este trabajo ha sido financiada por las siguientes ayudas: Beca pre-doctoral de formaci贸n de investigadores de la UAB (UAB2004-11), Fundaci贸n La Marat贸n de TV3, Fundaci贸n Alicia Koplowitz, Fundaci贸n Uriach y Ministerio Espa帽ol de Ciencia e Innovaci贸n (BFU2005-02783, BFU2008-04407).Over the years, numerous studies have demonstrated the fundamental role played by microglial cells in the CNS, not only in normal conditions where they control the correct tissue homeostasis, but also in all those situations in which, as a result of alterations and pathologies, homeostasis may be disturbed. Thus, when the integrity of the nervous tissue is disrupted, microglial cells are activated, showing specific activation patterns which fully depend on changes in the particular micro-environment. The microglial response has been widely studied in acute injuries in which microglia act as an intrinsic element of the innate immune system. However, the processes underlying microglial reactivity in situations of acquired immunity, as well as the relationship established between these microglial cells and infiltrating peripheral immune cells, remain poorly understood. In this study we have characterized the pattern of microglial reactivity and their relationship with the different populations of infiltrated lymphocytes, along the evolution of an acute model of experimental autoimmune encephalopathy (EAE) induced in Lewis rat. Our studies have demonstrated that microglial cells became activated in response to EAE induction, showing a specific activation pattern in each phase along disease evolution. During the inductive and peak phases, microglial cells showed changes in morphology and distribution, in close association with the increase of clinical symptoms, manifested by a progressive deterioration of motor function. These microglial cells progressively shorten their ramifications leading to amoeboid morphologies. Microglia and blood-borne macrophages increased in number and accumulated around blood vessels. In addition, a large number of infiltrated lymphocytes were also detected during the inductive and peak phases. These lymphocytes belong mostly to the T-helper phenotype (pro-inflammatory Th1 cells), although T-cytotoxic and γδ T-cells were also observed. Activated microglial cells displayed an immature dendritic cell phenotype characterized by expression of CD1 and major histocompatibility complexes class I and class II (MHC-class I and MHC-class II) without concomitant expression of B7.1 or B7.2 co-stimulatory molecules. In addition, these activated microglia expressed CD1, a marker of immature dendritic cells. In this context, the signal that these immature dendritic cell-like microglial cells induced to infiltrated lymphocytes may provoke lymphocyte apoptosis or anergy. During the recovery phase, animals experienced a gradual improvement in symptomatology, although microglial cells in this phase still showed a morphology, distribution and phenotype characteristics of reactive cells. In general, these microglial cells displayed the same immature dendritic cell phenotype (CD1+, MHC-I+, MHC-II+, B7.1- and B7.2-) observed during earlier phases. Noticeably, microglial cells located around blood vessels express B7.2. The total number of T-helper, T-cytotoxic and γδ T-cells remained very high with values close to those observed during the inductive and peak phases. Interestingly, we do not find Th1 lymphocytes during the recovery phase, and the population of T-helper cells mainly consists of Th17 and T-regs subtypes. Lymphocytes accumulated in the vicinity of blood vessels expressed CTLA-4, one of the B7.2 co卢receptors. In this new context, the interaction between B7.2+ microglial cells and CTLA-4+ lymphocytes could be responsible for the immune response resolution and the induction of subsequent tolerance. Altogether, these results show that microglia play a key role in the evolution of the acquired immune response, modulating the activation and inactivation of the different lymphocyte populations involved in both the induction of the immune/inflammatory process and its subsequent resolution. This work was supported by: UAB Pre-doctoral fellow (UAB2004-11), Fundaci贸n La Marat贸n de TV3, Fundaci贸n Alicia Koplowitz, Fundaci贸n Uriach y Ministerio Espa帽ol de Ciencia e Innovaci贸n (BFU2005-02783, BFU2008-04407)

    Are microglial cells the regulators of lymphocyte responses in the CNS?

    No full text
    The infiltration of immune cells in the central nervous system is a common hallmark in different neuroinflammatory conditions. Accumulating evidence indicates that resident glial cells can establish a cross-talk with infiltrated immune cells, including T-cells, regulating their recruitment, activation and function within the CNS. Although the healthy CNS has been thought to be devoid of professional dendritic cells (DCs), numerous studies have reported the presence of a population of DCs in specific locations such as the meninges, choroid plexuses and the perivascular space. Moreover, the infiltration of DC precursors during neuroinflammatory situations has been proposed, suggesting a putative role of these cells in the regulation of lymphocyte activity within the CNS. On the other hand, under specific circumstances, microglial cells are able to acquire a phenotype of DC expressing a wide range of molecules that equip these cells with all the necessary machinery for communication with T-cells. In this review, we summarize the current knowledge on the expression of molecules involved in the cross-talk with T-cells in both microglial cells and DCs and discuss the potential contribution of each of these cell populations on the control of lymphocyte function within the CNS

    Papel de la microgl铆a en la regulaci贸n de la respuesta inmunitaria adquirida

    Get PDF
    Descripci贸 del recurs: el 14 de febrer de 2011Numerosos estudios han demostrado a lo largo de los a帽os, el papel fundamental que juegan las c茅lulas de microgl铆a en el funcionamiento del SNC. No s贸lo en condiciones normales, donde controlan la correcta home贸stasis del tejido, sino tambi茅n en todas aquellas situaciones que, como consecuencia de alteraciones y procesos patol贸gicos diversos, conllevan a una p茅rdida de esta home贸stasis. En respuesta a todas estas situaciones, las c茅lulas de microgl铆a son capaces de detectar r谩pidamente el da帽o y actuar de una manera espec铆fica en funci贸n del tipo de perturbaci贸n que se produzca en su entorno. Esta respuesta microglial ha sido ampliamente estudiada en da帽os agudos, en los que la microgl铆a act煤a como parte del sistema inmune innato, sin embargo los procesos que subyacen a la reactividad microglial ante una situaci贸n de inmunidad adquirida, as铆 como la comunicaci贸n que se establece entre estas c茅lulas microgliales y las c茅lulas inmunes perif茅ricas infiltradas permanecen en muchos aspectos sin esclarecer. En el presente trabajo hemos caracterizado el patr贸n de reactividad microglial y su relaci贸n con las diferentes poblaciones de linfocitos infiltrados a lo largo de las diferentes fases de la evoluci贸n que acontecen en un modelo agudo de encefalopat铆a autoinmune experimental (EAE). Nuestros estudios demuestran que las c茅lulas de microgl铆a se activan en respuesta a la inducci贸n de la EAE y presentan un patr贸n de activaci贸n espec铆fico en cada una de las fases. Durante la fase de inducci贸n y pico del proceso patol贸gico, en estrecha relaci贸n con el aumento de la sintomatolog铆a cl铆nica que se manifiesta por un progresivo deterioro de las funciones motoras, estas c茅lulas microgliales experimentan cambios morfol贸gicos y en su distribuci贸n acumul谩ndose alrededor de los vasos sangu铆neos. Adem谩s de microgl铆a reactiva y posiblemente de macr贸fagos de origen sangu铆neo, se observa tambi茅n un gran n煤mero de linfocitos infiltrados, mayoritariamente del tipo T-cooperador y subtipo Th1 (pro-inflamatorio), si bien tambi茅n se observan linfocitos T-citot贸xicos y T-纬未. En estas fases, la activaci贸n microglial se caracteriza a nivel fenot铆pico por el aumento en la expresi贸n de mol茅culas del complejo mayor de histocompatibilidad clase I y clase II (MHC-clase I y MHC-clase II) sin expresi贸n concomitante de mol茅culas co-estimuladoras B7.1 o B7.2, es decir las c茅lulas de microgl铆a activadas presentan en estas circunstancias un fenotipo caracter铆stico de c茅lulas dendr铆ticas inmaduras; hecho que hemos corroborado con la demostraci贸n de la expresi贸n del marcador CD1. En este contexto, la se帽al que inducen estas c茅lulas de microgl铆a a los linfocitos infiltrados, podr铆a estar implicada en la modulaci贸n del proceso inflamatorio induciendo la apoptosis o anergia linfocitaria. Durante la fase de recuperaci贸n, a pesar de que los animales experimentan una progresiva mejora sintomatol贸gica, las c茅lulas de microgl铆a siguen mostrando una morfolog铆a, distribuci贸n y patr贸n de expresi贸n de mol茅culas caracter铆sticos de c茅lulas reactivas. Si bien, en general, las c茅lulas de microgl铆a siguen mostrando el mismo patr贸n fenot铆pico (CD1+, MHC-I+, MHC-II+, B7.1- y B7.2-), aquellas localizadas en el entorno de algunos vasos sangu铆neos expresan diferencialmente la mol茅cula B7.2. Durante esta fase, adem谩s, el n煤mero total de linfocitos T-cooperadores, T-citot贸xicos y T-纬未 se mantiene muy elevado con valores similares a los observados en las fases anteriores. Es interesante se帽alar que ya no encontramos linfocitos Th1 y la poblaci贸n de T-cooperadores est谩 constituida por los subtipos Th17 y T-regs. Los linfocitos que se acumulan en las inmediaciones de los vasos sangu铆neos expresan CTLA-4, uno de los co-receptores de B7.2. En este nuevo contexto, la interacci贸n de las c茅lulas de microgl铆a B7.2+ con estos linfocitos CTLA-4+ podr铆a ser la responsable de la resoluci贸n de la respuesta immunitaria y la inducci贸n de la tolerancia. En su conjunto los resultados obtenidos evidencian que la microgl铆a juega un papel clave en la evoluci贸n de la respuesta inmune adquirida modulando la activaci贸n e inactivaci贸n de las diferentes poblaciones linfocitarias implicadas tanto en la inducci贸n del proceso inmune/inflamatorio como en su posterior resoluci贸n. La realizaci贸n de este trabajo ha sido financiada por las siguientes ayudas: Beca pre-doctoral de formaci贸n de investigadores de la UAB (UAB2004-11), Fundaci贸n La Marat贸n de TV3, Fundaci贸n Alicia Koplowitz, Fundaci贸n Uriach y Ministerio Espa帽ol de Ciencia e Innovaci贸n (BFU2005-02783, BFU2008-04407).Over the years, numerous studies have demonstrated the fundamental role played by microglial cells in the CNS, not only in normal conditions where they control the correct tissue homeostasis, but also in all those situations in which, as a result of alterations and pathologies, homeostasis may be disturbed. Thus, when the integrity of the nervous tissue is disrupted, microglial cells are activated, showing specific activation patterns which fully depend on changes in the particular micro-environment. The microglial response has been widely studied in acute injuries in which microglia act as an intrinsic element of the innate immune system. However, the processes underlying microglial reactivity in situations of acquired immunity, as well as the relationship established between these microglial cells and infiltrating peripheral immune cells, remain poorly understood. In this study we have characterized the pattern of microglial reactivity and their relationship with the different populations of infiltrated lymphocytes, along the evolution of an acute model of experimental autoimmune encephalopathy (EAE) induced in Lewis rat. Our studies have demonstrated that microglial cells became activated in response to EAE induction, showing a specific activation pattern in each phase along disease evolution. During the inductive and peak phases, microglial cells showed changes in morphology and distribution, in close association with the increase of clinical symptoms, manifested by a progressive deterioration of motor function. These microglial cells progressively shorten their ramifications leading to amoeboid morphologies. Microglia and blood-borne macrophages increased in number and accumulated around blood vessels. In addition, a large number of infiltrated lymphocytes were also detected during the inductive and peak phases. These lymphocytes belong mostly to the T-helper phenotype (pro-inflammatory Th1 cells), although T-cytotoxic and 纬未 T-cells were also observed. Activated microglial cells displayed an immature dendritic cell phenotype characterized by expression of CD1 and major histocompatibility complexes class I and class II (MHC-class I and MHC-class II) without concomitant expression of B7.1 or B7.2 co-stimulatory molecules. In addition, these activated microglia expressed CD1, a marker of immature dendritic cells. In this context, the signal that these immature dendritic cell-like microglial cells induced to infiltrated lymphocytes may provoke lymphocyte apoptosis or anergy. During the recovery phase, animals experienced a gradual improvement in symptomatology, although microglial cells in this phase still showed a morphology, distribution and phenotype characteristics of reactive cells. In general, these microglial cells displayed the same immature dendritic cell phenotype (CD1+, MHC-I+, MHC-II+, B7.1- and B7.2-) observed during earlier phases. Noticeably, microglial cells located around blood vessels express B7.2. The total number of T-helper, T-cytotoxic and 纬未 T-cells remained very high with values close to those observed during the inductive and peak phases. Interestingly, we do not find Th1 lymphocytes during the recovery phase, and the population of T-helper cells mainly consists of Th17 and T-regs subtypes. Lymphocytes accumulated in the vicinity of blood vessels expressed CTLA-4, one of the B7.2 co卢receptors. In this new context, the interaction between B7.2+ microglial cells and CTLA-4+ lymphocytes could be responsible for the immune response resolution and the induction of subsequent tolerance. Altogether, these results show that microglia play a key role in the evolution of the acquired immune response, modulating the activation and inactivation of the different lymphocyte populations involved in both the induction of the immune/inflammatory process and its subsequent resolution. This work was supported by: UAB Pre-doctoral fellow (UAB2004-11), Fundaci贸n La Marat贸n de TV3, Fundaci贸n Alicia Koplowitz, Fundaci贸n Uriach y Ministerio Espa帽ol de Ciencia e Innovaci贸n (BFU2005-02783, BFU2008-04407)

    Increase in Th17 and T-reg lymphocytes and decrease of IL22 correlate with the recovery phase of acute EAE IN rat

    No full text
    Experimental autoimmune encephalomyelitis (EAE), a well-established model of multiple sclerosis, is characterised by microglial activation and lymphocyte infiltration. Induction of EAE in Lewis rats produces an acute monophasic disease characterised by a single peak of disability followed by a spontaneous and complete recovery and a subsequent tolerance to further immunizations. In the current study we have performed a detailed analysis of the dynamics of different lymphocyte populations and cytokine profile along the induction, peak, recovery and post-recovery phases in this paradigm. MBP-injected rats were sacrificed attending exclusively to their clinical score, and the different populations of T-lymphocytes as well as the dynamics of different pro- and anti-inflammatory cytokines were analysed in the spinal cord by flow cytometry, immunohistochemistry and ELISA. Our results revealed that, during the induction and peak phases, in parallel to an increase in symptomatology, the number of CD3+ and CD4+ cells increased progressively, showing a Th1 phenotype, but unexpectedly during recovery, although clinical signs progressively decreased, the number and proportion of CD3+ and CD4+ populations remained unaltered. Interestingly, during this recovery phase, we observed a marked decrease of Th1 and an important increase in Th17 and T-reg cells. Moreover, our results indicate a specific cytokine expression profile along the EAE course characterized by no changes of IL10 and IL17 levels, decrease of IL21 on the peak, and high IL22 levels during the induction and peak phases that markedly decrease during recovery. In summary, these results revealed the existence of a specific pattern of lymphocyte infiltration and cytokine secretion along the different phases of the acute EAE model in Lewis rat that differs from those already described in chronic or relapsing-remitting mouse models, where Th17-cells were found mostly during the peak, suggesting a specific role of these lymphocytes and cytokines in the evolution of this acute EAE model
    corecore