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












Base de datos
Intervalo de año de publicación
1.
Nature ; 627(8003): 399-406, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38448581

RESUMEN

Immune cells rely on transient physical interactions with other immune and non-immune populations to regulate their function1. To study these 'kiss-and-run' interactions directly in vivo, we previously developed LIPSTIC (labelling immune partnerships by SorTagging intercellular contacts)2, an approach that uses enzymatic transfer of a labelled substrate between the molecular partners CD40L and CD40 to label interacting cells. Reliance on this pathway limited the use of LIPSTIC to measuring interactions between CD4+ T helper cells and antigen-presenting cells, however. Here we report the development of a universal version of LIPSTIC (uLIPSTIC), which can record physical interactions both among immune cells and between immune and non-immune populations irrespective of the receptors and ligands involved. We show that uLIPSTIC can be used, among other things, to monitor the priming of CD8+ T cells by dendritic cells, reveal the steady-state cellular partners of regulatory T cells and identify germinal centre-resident T follicular helper cells on the basis of their ability to interact cognately with germinal centre B cells. By coupling uLIPSTIC with single-cell transcriptomics, we build a catalogue of the immune populations that physically interact with intestinal epithelial cells at the steady state and profile the evolution of the interactome of lymphocytic choriomeningitis virus-specific CD8+ T cells in multiple organs following systemic infection. Thus, uLIPSTIC provides a broadly useful technology for measuring and understanding cell-cell interactions across multiple biological systems.


Asunto(s)
Linfocitos B , Linfocitos T CD8-positivos , Comunicación Celular , Células Dendríticas , Células Epiteliales , Células T Auxiliares Foliculares , Linfocitos T Reguladores , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Ligandos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Células T Auxiliares Foliculares/citología , Células T Auxiliares Foliculares/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Centro Germinal/citología , Análisis de Expresión Génica de una Sola Célula , Células Epiteliales/citología , Células Epiteliales/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Especificidad de Órganos
2.
Cell ; 186(1): 131-146.e13, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36565697

RESUMEN

Germinal centers (GCs) form in secondary lymphoid organs in response to infection and immunization and are the source of affinity-matured B cells. The duration of GC reactions spans a wide range, and long-lasting GCs (LLGCs) are potentially a source of highly mutated B cells. We show that rather than consisting of continuously evolving B cell clones, LLGCs elicited by influenza virus or SARS-CoV-2 infection in mice are sustained by progressive replacement of founder clones by naive-derived invader B cells that do not detectably bind viral antigens. Rare founder clones that resist replacement for long periods are enriched in clones with heavily mutated immunoglobulins, including some with very high affinity for antigen, that can be recalled by boosting. Our findings reveal underappreciated aspects of the biology of LLGCs generated by respiratory virus infection and identify clonal replacement as a potential constraint on the development of highly mutated antibodies within these structures.


Asunto(s)
Linfocitos B , Centro Germinal , Infecciones por Virus ARN , Animales , Ratones , Linfocitos B/citología , Linfocitos B/inmunología , Células Clonales , COVID-19 , Centro Germinal/citología , Centro Germinal/inmunología , SARS-CoV-2 , Gripe Humana , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/patología , Infecciones por Virus ARN/virología
3.
Front Immunol ; 13: 1007080, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36451809

RESUMEN

Efficient mouse models to study SARS-CoV-2 infection are critical for the development and assessment of vaccines and therapeutic approaches to mitigate the current pandemic and prevent reemergence of COVID-19. While the first generation of mouse models allowed SARS-CoV-2 infection and pathogenesis, they relied on ectopic expression and non-physiological levels of human angiotensin-converting enzyme 2 (hACE2). Here we generated a mouse model carrying the minimal set of modifications necessary for productive infection with multiple strains of SARS-CoV-2. Substitution of only three amino acids in the otherwise native mouse Ace2 locus (Ace2 TripleMutant or Ace2™), was sufficient to render mice susceptible to both SARS-CoV-2 strains USA-WA1/2020 and B.1.1.529 (Omicron). Infected Ace2™ mice exhibited weight loss and lung damage and inflammation, similar to COVID-19 patients. Previous exposure to USA-WA1/2020 or mRNA vaccination generated memory B cells that participated in plasmablast responses during breakthrough B.1.1.529 infection. Thus, the Ace2™ mouse replicates human disease after SARS-CoV-2 infection and provides a tool to study immune responses to sequential infections in mice.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Ratones , Animales , Enzima Convertidora de Angiotensina 2/genética , Modelos Animales de Enfermedad , Pandemias
4.
Nature ; 610(7932): 547-554, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36198790

RESUMEN

Loss of Paneth cells and their antimicrobial granules compromises the intestinal epithelial barrier and is associated with Crohn's disease, a major type of inflammatory bowel disease1-7. Non-classical lymphoid cells, broadly referred to as intraepithelial lymphocytes (IELs), intercalate the intestinal epithelium8,9. This anatomical position has implicated them as first-line defenders in resistance to infections, but their role in inflammatory disease pathogenesis requires clarification. The identification of mediators that coordinate crosstalk between specific IEL and epithelial subsets could provide insight into intestinal barrier mechanisms in health and disease. Here we show that the subset of IELs that express γ and δ T cell receptor subunits (γδ IELs) promotes the viability of Paneth cells deficient in the Crohn's disease susceptibility gene ATG16L1. Using an ex vivo lymphocyte-epithelium co-culture system, we identified apoptosis inhibitor 5 (API5) as a Paneth cell-protective factor secreted by γδ IELs. In the Atg16l1-mutant mouse model, viral infection induced a loss of Paneth cells and enhanced susceptibility to intestinal injury by inhibiting the secretion of API5 from γδ IELs. Therapeutic administration of recombinant API5 protected Paneth cells in vivo in mice and ex vivo in human organoids with the ATG16L1 risk allele. Thus, we identify API5 as a protective γδ IEL effector that masks genetic susceptibility to Paneth cell death.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Enfermedad de Crohn , Predisposición Genética a la Enfermedad , Linfocitos Intraepiteliales , Proteínas Nucleares , Células de Paneth , Animales , Humanos , Ratones , Proteínas Reguladoras de la Apoptosis/metabolismo , Muerte Celular , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Predisposición Genética a la Enfermedad/genética , Mucosa Intestinal/patología , Proteínas Nucleares/metabolismo , Células de Paneth/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Supervivencia Celular , Organoides , Alelos
5.
Immunity ; 55(5): 800-818, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545029

RESUMEN

Pathogenic enteric viruses are a major cause of morbidity and mortality, particularly among children in developing countries. The host response to enteric viruses occurs primarily within the mucosa, where the intestinal immune system must balance protection against pathogens with tissue protection and tolerance to harmless commensal bacteria and food. Here, we summarize current knowledge in natural immunity to enteric viruses, highlighting specialized features of the intestinal immune system. We further discuss how knowledge of intestinal anti-viral mechanisms can be translated into vaccine development with particular focus on immunization in the oral route. Research reveals that the intestine is a complex interface between enteric viruses and the host where environmental factors influence susceptibility and immunity to infection, while viral infections can have lasting implications for host health. A deeper mechanistic understanding of enteric anti-viral immunity with this broader context can ultimately lead to better vaccines for existing and emerging viruses.


Asunto(s)
Infecciones por Enterovirus , Vacunas , Virus , Antígenos Virales , Niño , Humanos , Inmunidad Innata , Mucosa Intestinal , Intestinos
6.
Immunity ; 55(7): 1234-1249.e6, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35617965

RESUMEN

The intestinal epithelium comprises the body's largest surface exposed to viruses. Additionally, the gut epithelium hosts a large population of intraepithelial T lymphocytes, or IELs, although their role in resistance against viral infections remains elusive. By fate-mapping T cells recruited to the murine intestine, we observed an accumulation of newly recruited CD4+ T cells after infection with murine norovirus CR6 and adenovirus type-2 (AdV), but not reovirus. CR6- and AdV-recruited intraepithelial CD4+ T cells co-expressed Ly6A and chemokine receptor CCR9, exhibited T helper 1 and cytotoxic profiles, and conferred protection against AdV in vivo and in an organoid model in an IFN-γ-dependent manner. Ablation of the T cell receptor (TCR) or the transcription factor ThPOK in CD4+ T cells prior to AdV infection prevented viral control, while TCR ablation during infection did not impact viral clearance. These results uncover a protective role for intraepithelial Ly6A+CCR9+CD4+ T cells against enteric adenovirus.


Asunto(s)
Intestino Delgado , Virosis , Animales , Antígenos Ly , Linfocitos T CD4-Positivos , Mucosa Intestinal , Proteínas de la Membrana , Ratones , Receptores de Quimiocina
7.
Science ; 371(6535)2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33737460

RESUMEN

The intestine is a site of direct encounter with the external environment and must consequently balance barrier defense with nutrient uptake. To investigate how nutrient uptake is regulated in the small intestine, we tested the effect of diets with different macronutrient compositions on epithelial gene expression. We found that enzymes and transporters required for carbohydrate digestion and absorption were regulated by carbohydrate availability. The "on-demand" induction of this machinery required γδ T cells, which regulated this program through the suppression of interleukin-22 production by type 3 innate lymphoid cells. Nutrient availability altered the tissue localization and transcriptome of γδ T cells. Additionally, transcriptional responses to diet involved cellular remodeling of the epithelial compartment. Thus, this work identifies a role for γδ T cells in nutrient sensing.


Asunto(s)
Carbohidratos de la Dieta/administración & dosificación , Carbohidratos de la Dieta/metabolismo , Enterocitos/fisiología , Interleucinas/metabolismo , Mucosa Intestinal/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta , Subgrupos de Linfocitos T/fisiología , Adaptación Fisiológica , Animales , Comunicación Celular , Proteínas en la Dieta/administración & dosificación , Digestión , Regulación de la Expresión Génica , Interleucinas/genética , Absorción Intestinal , Mucosa Intestinal/citología , Intestino Delgado/citología , Intestino Delgado/metabolismo , Ratones Endogámicos C57BL , Nutrientes/administración & dosificación , Nutrientes/metabolismo , Subgrupos de Linfocitos T/inmunología , Transcripción Genética , Transcriptoma , Interleucina-22
8.
Sci Immunol ; 5(52)2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33067381

RESUMEN

Multiple sclerosis (MS) is a leading cause of incurable progressive disability in young adults caused by inflammation and neurodegeneration in the central nervous system (CNS). The capacity of microglia to clear tissue debris is essential for maintaining and restoring CNS homeostasis. This capacity diminishes with age, and age strongly associates with MS disease progression, although the underlying mechanisms are still largely elusive. Here, we demonstrate that the recovery from CNS inflammation in a murine model of MS is dependent on the ability of microglia to clear tissue debris. Microglia-specific deletion of the autophagy regulator Atg7, but not the canonical macroautophagy protein Ulk1, led to increased intracellular accumulation of phagocytosed myelin and progressive MS-like disease. This impairment correlated with a microglial phenotype previously associated with neurodegenerative pathologies. Moreover, Atg7-deficient microglia showed notable transcriptional and functional similarities to microglia from aged wild-type mice that were also unable to clear myelin and recover from disease. In contrast, induction of autophagy in aged mice using the disaccharide trehalose found in plants and fungi led to functional myelin clearance and disease remission. Our results demonstrate that a noncanonical form of autophagy in microglia is responsible for myelin degradation and clearance leading to recovery from MS-like disease and that boosting this process has a therapeutic potential for age-related neuroinflammatory conditions.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/deficiencia , Encefalomielitis Autoinmune Experimental/inmunología , Microglía/inmunología , Esclerosis Múltiple/inmunología , Fagocitosis/inmunología , Animales , Autofagia/inmunología , Proteína 7 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/deficiencia , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Encéfalo/citología , Encéfalo/inmunología , Encéfalo/patología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Microglía/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo , Cultivo Primario de Células , Médula Espinal/citología , Médula Espinal/inmunología , Médula Espinal/patología
9.
Nat Commun ; 9(1): 4845, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451869

RESUMEN

Circulating monocytes can compete for virtually any tissue macrophage niche and become long-lived replacements that are phenotypically indistinguishable from their embryonic counterparts. As the factors regulating this process are incompletely understood, we studied niche competition in the brain by depleting microglia with >95% efficiency using Cx3cr1CreER/+R26DTA/+ mice and monitored long-term repopulation. Here we show that the microglial niche is repopulated within weeks by a combination of local proliferation of CX3CR1+F4/80lowClec12a- microglia and infiltration of CX3CR1+F4/80hiClec12a+ macrophages that arise directly from Ly6Chi monocytes. This colonization is independent of blood brain barrier breakdown, paralleled by vascular activation, and regulated by type I interferon. Ly6Chi monocytes upregulate microglia gene expression and adopt microglia DNA methylation signatures, but retain a distinct gene signature from proliferating microglia, displaying altered surface marker expression, phagocytic capacity and cytokine production. Our results demonstrate that monocytes are imprinted by the CNS microenvironment but remain transcriptionally, epigenetically and functionally distinct.


Asunto(s)
Encéfalo/inmunología , Linaje de la Célula/inmunología , Regulación de la Expresión Génica/inmunología , Microglía/inmunología , Monocitos/inmunología , Traslado Adoptivo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos Ly/genética , Antígenos Ly/inmunología , Proteínas Bacterianas/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Encéfalo/citología , Encéfalo/efectos de la radiación , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Linaje de la Célula/efectos de la radiación , Proliferación Celular , Metilación de ADN , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Proteínas Luminiscentes/inmunología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/efectos de la radiación , Ratones , Ratones Transgénicos , Microglía/citología , Microglía/efectos de la radiación , Monocitos/citología , Monocitos/efectos de la radiación , Monocitos/trasplante , Fagocitosis , Receptores Mitogénicos/genética , Receptores Mitogénicos/inmunología , Transducción de Señal , Quimera por Trasplante , Irradiación Corporal Total
10.
Nat Immunol ; 19(5): 1-7, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29662171

RESUMEN

The cytokine transforming growth factor-ß (TGF-ß) regulates the development and homeostasis of several tissue-resident macrophage populations, including microglia. TGF-ß is not critical for microglia survival but is required for the maintenance of the microglia-specific homeostatic gene signature1,2. Under defined host conditions, circulating monocytes can compete for the microglial niche and give rise to long-lived monocyte-derived macrophages residing in the central nervous system (CNS)3-5. Whether monocytes require TGF-ß for colonization of the microglial niche and maintenance of CNS integrity is unknown. We found that abrogation of TGF-ß signaling in CX3CR1+ monocyte-derived macrophages led to rapid onset of a progressive and fatal demyelinating motor disease characterized by myelin-laden giant macrophages throughout the spinal cord. Tgfbr2-deficient macrophages were characterized by high expression of genes encoding proteins involved in antigen presentation, inflammation and phagocytosis. TGF-ß is thus crucial for the functional integration of monocytes into the CNS microenvironment.


Asunto(s)
Encéfalo/inmunología , Enfermedades Desmielinizantes/inmunología , Macrófagos/patología , Médula Espinal/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Transducción de Señal , Médula Espinal/metabolismo , Médula Espinal/patología , Factor de Crecimiento Transformador beta/metabolismo
11.
Nat Immunol ; 17(12): 1407-1414, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27798616

RESUMEN

The innate responsiveness of the immune system is important not only for quick responses to pathogens but also for the initiation and shaping of the subsequent adaptive immune response. Activation via the cytokine IL-18, a product of inflammasomes, gives rise to a rapid response that includes the production of self-reactive antibodies. As increased concentrations of this cytokine are found in inflammatory diseases, we investigated the origin of the B cell response and its regulation. We identified an accumulation of B cell-helper neutrophils in the spleen that interacted with innate-type invariant natural killer T cells (iNKT cells) to regulate B cell responses. We found that neutrophil-dependent expression of the death-receptor ligand FasL by iNKT cells was needed to restrict autoantibody production. Neutrophils can thus license iNKT cells to regulate potentially harmful autoreactive B cell responses during inflammasome-driven inflammation.


Asunto(s)
Linfocitos B/inmunología , Proteína Ligando Fas/metabolismo , Inflamación/inmunología , Células T Asesinas Naturales/inmunología , Neutrófilos/inmunología , Inmunidad Adaptativa , Animales , Autoanticuerpos/biosíntesis , Células Cultivadas , Proteína Ligando Fas/genética , Inmunidad Innata , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Glia ; 64(11): 1925-37, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27479807

RESUMEN

Intracerebral levels of Transforming Growth Factor beta (TGFß) rise rapidly during the onset of experimental autoimmune encephalomyelitis (EAE), a mouse model of Multiple Sclerosis (MS). We addressed the role of TGFß responsiveness in EAE by targeting the TGFß receptor in myeloid cells, determining that Tgfbr2 was specifically targeted in monocyte-derived dendritic cells (moDCs) but not in CNS resident microglia by using bone-marrow chimeric mice. TGFß responsiveness in moDCs was necessary for the remission phase since LysM(Cre) Tgfbr2(fl/fl) mice developed a chronic form of EAE characterized by severe demyelination and extensive infiltration of activated moDCs in the CNS. Tgfbr2 deficiency resulted in increased moDC IL-12 secretion that skewed T cells to produce IFN-γ, which in turn enhanced the production of moDC-derived reactive oxygen species that promote oxidative damage and demyelination. We identified SNPs in the human NOX2 (CYBB) gene that associated with the severity of MS, and significantly increased CYBB expression was recorded in PBMCs from both MS patients and from MS severity risk allele rs72619425-A carrying individuals. We thus identify a novel myeloid cell-T cell activation loop active in the CNS during chronic disease that could be therapeutically targeted. GLIA 2016;64:1925-1937.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Polaridad Celular/fisiología , Citocinas/metabolismo , Células Dendríticas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Células TH1/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Polaridad Celular/genética , Estudios de Cohortes , Citocinas/genética , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica/genética , Genotipo , Humanos , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/citología , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo , Polimorfismo de Nucleótido Simple/genética , Factor de Crecimiento Transformador beta/genética
13.
J Exp Med ; 213(8): 1537-53, 2016 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-27432941

RESUMEN

Prolonged infections or adjuvant usage can trigger emergency granulopoiesis (EG), leading to dysregulation in neutrophil blood counts. However, the impact of EG on T and B cell function remains largely unknown. In this study, to address this question, we used a mouse model of neutropenia and studied immune activation after adjuvant administration. The initial neutropenic state fostered an environment of increased dendritic cell activation and T cell-derived IL-17 production. Interestingly, neutropenic lysozyme 2-diphtheria toxin A mice exhibited striking EG and amplified neutrophil recruitment to the lymph nodes (LNs) that was dependent on IL-17-induced prostaglandin activity. The recruited neutrophils secreted a B cell-activating factor that highly accelerated plasma cell generation and antigen-specific antibody production. Reduction of neutrophil functions via granulocyte colony-stimulating factor neutralization significantly diminished plasma cell formation, directly linking EG with the humoral immune response. We conclude that neutrophils are capable of directly regulating T cell-dependent B cell responses in the LN.


Asunto(s)
Formación de Anticuerpos/fisiología , Factor Activador de Células B/metabolismo , Inmunidad Humoral/fisiología , Mielopoyesis/fisiología , Neutrófilos/metabolismo , Células Plasmáticas/metabolismo , Animales , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-17/metabolismo , Ratones , Ratones Noqueados , Neutropenia/genética , Neutropenia/inmunología , Neutropenia/metabolismo , Neutrófilos/citología , Neutrófilos/inmunología , Células Plasmáticas/inmunología
14.
Glia ; 62(5): 804-17, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24677019

RESUMEN

Microglia are resident antigen-presenting cells in the central nervous system (CNS) that either suppress or promote disease depending on their activation phenotype and the microenvironment. Multiple sclerosis (MS) is a chronic inflammatory disease causing demyelination and nerve loss in the CNS, and experimental autoimmune encephalomyelitis (EAE) is an animal model of MS that is widely used to investigate pathogenic mechanisms and therapeutic effects. We isolated and cultured microglia from adult mouse brains and exposed them to specific combinations of stimulatory molecules and cytokines, the combination of IL-4, IL-10, and TGF-ß yielding the optimal regime for induction of an immunosuppressive phenotype (M2). M2 microglia were characterized by decreased expression or production of CD86, PD-L1, nitric oxide, and IL-6, increased expression of PD-L2, and having a potent capacity to retain their phenotype on secondary proinflammatory stimulation. M2 microglia induced regulatory T cells, suppressed T-cell proliferation, and downmodulated M1-associated receptor expression in M1 macrophages. Myelin oligodendrocyte glycoprotein (MOG)-induced EAE was induced in DBA/1 mice and at different time points (0, 5, 12, or 15 days postimmunization) 3 × 105 M2 microglia were transferred intranasally. A single transfer of M2 microglia attenuated the severity of established EAE, which was particularly obvious when the cells were injected at 15 days postimmunization. M2 microglia-treated mice had reduced inflammatory responses and less demyelination in the CNS. Our findings demonstrate that adult M2 microglia therapy represents a novel intervention that alleviated established EAE and that this therapeutic principle may have relevance for treatment of MS patients.


Asunto(s)
Traslado Adoptivo/métodos , Citocinas/farmacología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Microglía/inmunología , Microglía/trasplante , Animales , Técnicas de Cocultivo , Femenino , Humanos , Inmunomodulación/inmunología , Ratones , Ratones Endogámicos DBA , Microglía/efectos de los fármacos
15.
PLoS One ; 8(12): e81912, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24312603

RESUMEN

Defining genetic variants that predispose for diseases is an important initiative that can improve biological understanding and focus therapeutic development. Genetic mapping in humans and animal models has defined genomic regions controlling a variety of phenotypes known as quantitative trait loci (QTL). Causative disease determinants, including single nucleotide polymorphisms (SNPs), lie within these regions and can often be identified through effects on gene expression. We previously identified a QTL on rat chromosome 4 regulating macrophage phenotypes and immune-mediated diseases including experimental autoimmune encephalomyelitis (EAE). Gene analysis and a literature search identified lysine-specific demethylase 3A (Kdm3a) as a potential regulator of these phenotypes. Genomic sequencing determined only two synonymous SNPs in Kdm3a. The silent synonymous SNP in exon 15 of Kdm3a caused problems with quantitative PCR detection in the susceptible strain through reduced amplification efficiency due to altered secondary cDNA structure. Shape Probability Shift analysis predicted that the SNP often affects RNA folding; thus, it may impact protein translation. Despite these differences in rats, genetic knockout of Kdm3a in mice resulted in no dramatic effect on immune system development and activation or EAE susceptibility and severity. These results provide support for tools that analyze causative SNPs that impact nucleic acid structures.


Asunto(s)
ADN/química , Exones/genética , Silenciador del Gen , Histona Demetilasas con Dominio de Jumonji/deficiencia , Histona Demetilasas con Dominio de Jumonji/genética , Polimorfismo de Nucleótido Simple/genética , ARN/química , Animales , Secuencia de Bases , ADN Complementario/genética , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Técnicas de Silenciamiento del Gen , Ratones , Fenotipo , Ratas
16.
J Neuroinflammation ; 10: 60, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23656637

RESUMEN

BACKGROUND: C-type lectin (CLEC) receptors are important for initiating and shaping immune responses; however, their role in inflammatory reactions in the central nervous system after traumatic injuries is not known. The antigen-presenting lectin-like receptor gene complex (Aplec) contains a few CLEC genes, which differ genetically among inbred rat strains. It was originally thought to be a region that regulates susceptibility to autoimmune arthritis, autoimmune neuroinflammation and infection. METHODS: The inbred rat strains DA and PVG differ substantially in degree of spinal cord motor neuron death following ventral root avulsion (VRA), which is a reproducible model of localized nerve root injury. A large F2 (DAxPVG) intercross was bred and genotyped after which global expressional profiling was performed on spinal cords from F2 rats subjected to VRA. A congenic strain, Aplec, created by transferring a small PVG segment containing only seven genes, all C-type lectins, ontoDA background, was used for further experiments together with the parental strains. RESULTS: Global expressional profiling of F2 (DAxPVG) spinal cords after VRA and genome-wide eQTL mapping identified a strong cis-regulated difference in the expression of Clec4a3 (Dcir3), a C-type lectin gene that is a part of the Aplec cluster. Second, we demonstrate significantly improved motor neuron survival and also increased T-cell infiltration into the spinal cord of congenic rats carrying Aplec from PVG on DA background compared to the parental DA strain. In vitro studies demonstrate that the Aplec genes are expressed on microglia and upregulated upon inflammatory stimuli. However, there were no differences in expression of general microglial activation markers between Aplec and parental DA rats, suggesting that the Aplec genes are involved in the signaling events rather than the primary activation of microglia occurring upon nerve root injury. CONCLUSIONS: In summary, we demonstrate that a genetic variation in Aplec occurring among inbred strains regulates both survival of axotomized motor neurons and the degree of lymphocyte infiltration. These results demonstrate a hitherto unknown role for CLECs for intercellular communication that occurs after damage to the nervous system, which is relevant for neuronal survival.


Asunto(s)
Lectinas Tipo C/genética , Neuronas Motoras/fisiología , Familia de Multigenes/genética , Radiculopatía/genética , Radiculopatía/patología , Linfocitos T/fisiología , Animales , Animales Congénicos , Presentación de Antígeno , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Astrocitos/metabolismo , Recuento de Células , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Citometría de Flujo , Inmunohistoquímica , Lectinas Tipo C/metabolismo , Análisis por Micromatrices , Microglía/metabolismo , Proteínas de la Mielina/metabolismo , Oligodendroglía/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Raíces Nerviosas Espinales/patología
17.
Brain Behav Immun ; 27(1): 109-22, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23044177

RESUMEN

Increasing evidence suggests that genetic background affects outcome of traumatic brain injuries (TBI). Still, there is limited detailed knowledge on what pathways/processes are affected by genetic heterogeneity. The inbred rat strains DA and PVG differ in neuronal survival following TBI. We here carried out global expressional profiling to identify differentially regulated pathways governing the response to an experimental controlled brain contusion injury. One of the most differentially regulated molecular networks concerned immune cell trafficking. Subsequent characterization of the involved cells using flow cytometry demonstrated greater infiltration of neutrophils and monocytes, as well as a higher degree of microglia activation in DA compared to PVG rats. In addition, DA rats displayed a higher number of NK cells and a higher ratio of CD161bright compared to CD161dim NK cells. Local expression of complement pathway molecules such as C1 and C3 was higher in DA and both the key complement component C3 and membrane-attack complex (MAC) could be demonstrated on axons and nerve cells. A stronger activation of the complement system in DA was associated with higher cerebrospinal fluid levels of neurofilament-light, a biomarker for nerve/axonal injury. In summary, we demonstrate substantial differences between DA and PVG rats in activation of inflammatory pathways; in particular, immune cell influx and complement activation associated with neuronal/axonal injury after TBI. These findings suggest genetic influences acting on inflammatory activation to be of importance in TBI and motivate further efforts using experimental forward genetics to identify genes/pathways that affect outcome.


Asunto(s)
Lesiones Encefálicas , Activación de Complemento , Leucocitos , ARN Mensajero/análisis , Ratas Endogámicas , Animales , Lesiones Encefálicas/genética , Lesiones Encefálicas/inmunología , Movimiento Celular/genética , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C1q/genética , Complemento C1q/inmunología , Complemento C3/genética , Complemento C3/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/inmunología , Citocinas/genética , Citocinas/inmunología , Perfilación de la Expresión Génica , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Leucocitos/citología , Leucocitos/inmunología , Microglía/citología , Microglía/inmunología , Monocitos/citología , Monocitos/inmunología , Subfamilia B de Receptores Similares a Lectina de Células NK/genética , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Neutrófilos/citología , Neutrófilos/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Endogámicas/genética , Ratas Endogámicas/inmunología
18.
Glia ; 60(12): 1930-43, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22911652

RESUMEN

The sentinel and immune functions of microglia require rapid and appropriate reactions to infection and damage. Their Toll-like receptors (TLRs) sense both as threats. However, whether activated microglia mount uniform responses or whether subsets conduct selective tasks is unknown. We demonstrate that murine microglia reorganize their responses to TLR activations postnatally and that this process comes with a maturation of TLR4-organized functions. Although induction of MHCI for antigen presentation remains as a pan-populational feature, synthesis of TNFα becomes restricted to a subset, even within adult central nervous system regions. Response heterogeneity is evident ex vivo, in situ, and in vivo, but is not limited to TNFα production or to TLR-triggered functions. Also, clearance activities for myelin under physiological and pathophysiological conditions, IFNγ-enforced upregulation of MHCII, or challenged inductions of other proinflammatory factors reveal dissimilar microglial contributions. Notably, response heterogeneity is also confirmed in human brain tissue. Our findings suggest that microglia divide by constitutive and inducible capacities. Privileged production of inflammatory mediators assigns a master control to subsets. Sequestration of clearance of endogenous material versus antigen presentation in exclusive compartments can separate potentially interfering functions. Finally, subsets rather than a uniform population of microglia may assemble the reactive phenotypes in responses during infection, injury, and rebuilding, warranting consideration in experimental manipulation and therapeutic strategies.


Asunto(s)
Microglía/clasificación , Microglía/metabolismo , Receptor Toll-Like 4/metabolismo , Anciano , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Receptor Toll-Like 4/fisiología
19.
Diabetes ; 61(11): 2881-92, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22745325

RESUMEN

Macrophages are multifunctional immune cells that may either drive or modulate disease pathogenesis depending on their activation phenotype. Autoimmune type 1 diabetes (T1D) is a chronic proinflammatory condition characterized by unresolved destruction of pancreatic islets. Adoptive cell transfer of macrophages with immunosuppressive properties represents a novel immunotherapy for treatment of such chronic autoimmune diseases. We used a panel of cytokines and other stimuli to discern the most effective regimen for in vitro induction of immunosuppressive macrophages (M2r) and determined interleukin (IL)-4/IL-10/transforming growth factor-ß (TGF-ß) to be optimal. M2r cells expressed programmed cell death 1 ligand-2, fragment crystallizable region γ receptor IIb, IL-10, and TGF-ß, had a potent deactivating effect on proinflammatory lipopolysaccharide/interferon-γ-stimulated macrophages, and significantly suppressed T-cell proliferation. Clinical therapeutic efficacy was assessed after adoptive transfer in NOD T1D mice, and after a single transfer of M2r macrophages, >80% of treated NOD mice were protected against T1D for at least 3 months, even when transfer was conducted just prior to clinical onset. Fluorescent imaging analyses revealed that adoptively transferred M2r macrophages specifically homed to the inflamed pancreas, promoting ß-cell survival. We suggest that M2r macrophage therapy represents a novel intervention that stops ongoing autoimmune T1D and may have relevance in a clinical setting.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Inmunoterapia Adoptiva , Células Secretoras de Insulina/inmunología , Macrófagos/inmunología , Macrófagos/trasplante , Animales , Proliferación Celular , Rastreo Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Femenino , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Ensayos de Liberación de Interferón gamma , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Activación de Macrófagos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Páncreas/inmunología , Páncreas/metabolismo , Páncreas/patología , Estado Prediabético/inmunología , Estado Prediabético/metabolismo , Estado Prediabético/patología , Estado Prediabético/terapia , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
20.
J Immunol ; 182(5): 3105-11, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19234207

RESUMEN

We recently reported that administration of tumor-specific bacteriophages initiates infiltration of neutrophilic granulocytes with subsequent regression of established B16 tumors. The aim of the current study was to investigate the mechanism of action of bacteriophage-induced tumor regression and to examine possible stimulatory effects of bacteriophages on macrophages. We observed that the mechanism of phage-induced tumor regression is TLR dependent as no signs of tumor destruction or neutrophil infiltration were observed in tumors in MyD88(-/-) mice in which TLR signaling is abolished. The microenvironment of bacteriophage-treated tumors was further analyzed by gene profiling through applying a low-density array preferentially designed to detect genes expressed by activated APCs, which demonstrated that the M2-polarized tumor microenvironment switched to a more M1-polarized milieu following phage treatment. Bacteriophage stimulation induced secretion of proinflammatory cytokines in both normal mouse macrophages and tumor-associated macrophages (TAMs) and increased expression of molecules involved in Ag presentation and costimulation. Furthermore, mouse neutrophils selectively migrated toward mediators secreted by bacteriophage-stimulated TAMs. Under these conditions, the neutrophils also exhibited increased cytotoxicity toward B16 mouse melanoma target cells. These results describe a close interplay of the innate immune system in which bacteriophages, located to the tumor microenvironment due to their specificity, stimulate TAMs to secrete factors that promote recruitment of neutrophils and potentiate neutrophil-mediated tumor destruction.


Asunto(s)
Bacteriófago M13/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/patología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Apoptosis/inmunología , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Femenino , Macrófagos/virología , Melanoma Experimental/patología , Melanoma Experimental/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Receptores Toll-Like/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...