Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 21(1): 71, 2024 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-38521932

RESUMEN

Cerebrospinal fluid (CSF) matrix biomarkers have become increasingly valuable surrogate markers of neuropsychiatric diseases in research and clinical practice. In contrast, CSF cells have been rarely investigated due to their relative scarcity and fragility, and lack of common collection and cryopreservation protocols, with limited exceptions for neurooncology and primary immune-based diseases like multiple sclerosis. the advent of a microfluidics-based multi-omics approach to studying individual cells has allowed for the study of cellular phenotyping, intracellular dynamics, and intercellular relationships that provide multidimensionality unable to be obtained through acellular fluid-phase analyses. challenges to cell-based research include site-to-site differences in handling, storage, and thawing methods, which can lead to inaccuracy and inter-assay variability. In the present study, we performed single-cell RNA sequencing (10x Genomics) on fresh or previously cryopreserved human CSF samples from three alternative cryopreservation methods: Fetal Bovine Serum with Dimethyl sulfoxide (FBS/DMSO), FBS/DMSO after a DNase step (a step often included in epigenetic studies), and cryopreservation using commercially available Recovery© media. In comparing relative differences between fresh and cryopreserved samples, we found little effect of the cryopreservation method on being able to resolve donor-linked cell type proportions, markers of cellular stress, and overall gene expression at the single-cell level, whereas donor-specific differences were readily discernable. We further demonstrate the compatibility of fresh and cryopreserved CSF immune cell sequencing using biologically relevant sexually dimorphic gene expression differences by donor. Our findings support the utility and interchangeability of FBS/DMSO and Recovery cryopreservation with fresh sample analysis, providing a methodological grounding that will enable researchers to further expand our understanding of the CSF immune cell contributions to neurological and psychiatric disease.


Asunto(s)
Crioprotectores , Dimetilsulfóxido , Humanos , Dimetilsulfóxido/farmacología , Crioprotectores/farmacología , Células Cultivadas , Criopreservación/métodos , Análisis de la Célula Individual , Supervivencia Celular
2.
Proc Natl Acad Sci U S A ; 120(32): e2306731120, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37523555

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal disease affecting upper and lower motor neurons. Microglia directly interact with motor neurons and participate in the progression of ALS. Single-cell mass cytometry (CyTOF) analysis revealed prominent expression of α5 integrin in microglia and macrophages in a superoxide dismutase-1 G93A mouse model of ALS (SOD1G93A). In postmortem tissues from ALS patients with various clinical ALS phenotypes and disease duration, α5 integrin is prominent in motor pathways of the central and peripheral nervous system and in perivascular zones associated with the blood-brain barrier. In SOD1G93A mice, administration of a monoclonal antibody against α5 integrin increased survival compared to an isotype control and improved motor function on behavioral testing. Together, these findings in mice and in humans suggest that α5 integrin is a potential therapeutic target in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Corteza Motora , Ratones , Humanos , Animales , Esclerosis Amiotrófica Lateral/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Integrina alfa5/metabolismo , Ratones Transgénicos , Superóxido Dismutasa/metabolismo , Macrófagos/metabolismo , Modelos Animales de Enfermedad
3.
Neuron ; 110(21): 3458-3483, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36327895

RESUMEN

Microglial research has advanced considerably in recent decades yet has been constrained by a rolling series of dichotomies such as "resting versus activated" and "M1 versus M2." This dualistic classification of good or bad microglia is inconsistent with the wide repertoire of microglial states and functions in development, plasticity, aging, and diseases that were elucidated in recent years. New designations continuously arising in an attempt to describe the different microglial states, notably defined using transcriptomics and proteomics, may easily lead to a misleading, although unintentional, coupling of categories and functions. To address these issues, we assembled a group of multidisciplinary experts to discuss our current understanding of microglial states as a dynamic concept and the importance of addressing microglial function. Here, we provide a conceptual framework and recommendations on the use of microglial nomenclature for researchers, reviewers, and editors, which will serve as the foundations for a future white paper.


Asunto(s)
Microglía
5.
Biomedicines ; 10(4)2022 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-35453590

RESUMEN

The innate immune landscape of the central nervous system (CNS), including the brain and the retina, consists of different myeloid cell populations with distinct tasks to fulfill. Whereas the CNS borders harbor extraparenchymal CNS-associated macrophages whose main duty is to build up a defense against invading pathogens and other damaging factors from the periphery, the resident immune cells of the CNS parenchyma and the retina, microglia, are highly dynamic cells with a plethora of functions during homeostasis and disease. Therefore, microglia are constantly sensing their environment and closely interacting with surrounding cells, which is in part mediated by soluble factors. One of these factors is Osteopontin (OPN), a multifunctional protein that is produced by different cell types in the CNS, including microglia, and is upregulated in neurodegenerative and neuroinflammatory conditions. In this review, we discuss the current literature about the interaction between microglia and OPN in homeostasis and several disease entities, including multiple sclerosis (MS), Alzheimer's and cerebrovascular diseases (AD, CVD), amyotrophic lateral sclerosis (ALS), age-related macular degeneration (AMD) and diabetic retinopathy (DR), in the context of the molecular pathways involved in OPN signaling shaping the function of microglia. As nearly all CNS diseases are characterized by pathological alterations in microglial cells, accompanied by the disturbance of the homeostatic microglia phenotype, the emergence of disease-associated microglia (DAM) states and their interplay with factors shaping the DAM-signature, such as OPN, is of great interest for therapeutical interventions in the future.

6.
Nat Neurosci ; 21(4): 541-551, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29507414

RESUMEN

Neuroinflammation and neurodegeneration may represent two poles of brain pathology. Brain myeloid cells, particularly microglia, play key roles in these conditions. We employed single-cell mass cytometry (CyTOF) to compare myeloid cell populations in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis, the R6/2 model of Huntington's disease (HD) and the mutant superoxide dismutase 1 (mSOD1) model of amyotrophic lateral sclerosis (ALS). We identified three myeloid cell populations exclusive to the CNS and present in each disease model. Blood-derived monocytes comprised five populations and migrated to the brain in EAE, but not in HD and ALS models. Single-cell analysis resolved differences in signaling and cytokine production within similar myeloid populations in EAE compared to HD and ALS models. Moreover, these analyses highlighted α5 integrin on myeloid cells as a potential therapeutic target for neuroinflammation. Together, these findings illustrate how neuropathology may differ between inflammatory and degenerative brain disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Encéfalo/patología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Enfermedad de Huntington/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Mieloides/patología , Animales , Proteína de Unión a CREB/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Monocitos , Mutación/genética , Células Mieloides/metabolismo , Análisis de la Célula Individual/métodos , Superóxido Dismutasa-1/genética
8.
Proc Natl Acad Sci U S A ; 113(12): E1738-46, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26884166

RESUMEN

The specific function of microglia, the tissue resident macrophages of the brain and spinal cord, has been difficult to ascertain because of a lack of tools to distinguish microglia from other immune cells, thereby limiting specific immunostaining, purification, and manipulation. Because of their unique developmental origins and predicted functions, the distinction of microglia from other myeloid cells is critically important for understanding brain development and disease; better tools would greatly facilitate studies of microglia function in the developing, adult, and injured CNS. Here, we identify transmembrane protein 119 (Tmem119), a cell-surface protein of unknown function, as a highly expressed microglia-specific marker in both mouse and human. We developed monoclonal antibodies to its intracellular and extracellular domains that enable the immunostaining of microglia in histological sections in healthy and diseased brains, as well as isolation of pure nonactivated microglia by FACS. Using our antibodies, we provide, to our knowledge, the first RNAseq profiles of highly pure mouse microglia during development and after an immune challenge. We used these to demonstrate that mouse microglia mature by the second postnatal week and to predict novel microglial functions. Together, we anticipate these resources will be valuable for the future study and understanding of microglia in health and disease.


Asunto(s)
Encéfalo/citología , Proteínas de la Membrana/análisis , Microglía/química , Proteínas del Tejido Nervioso/análisis , Anciano , Animales , Anticuerpos Monoclonales/inmunología , Biomarcadores , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , División Celular , Linaje de la Célula , Niño , Endotoxemia/patología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Lipopolisacáridos/toxicidad , Macrófagos/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Microglía/fisiología , Persona de Mediana Edad , Compresión Nerviosa , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Traumatismos del Nervio Óptico/patología , Especificidad de Órganos , Conejos , Nervio Ciático/lesiones , Nervio Ciático/patología , Análisis de Secuencia de ARN , Lóbulo Temporal/metabolismo , Transcriptoma
9.
Arthritis Rheumatol ; 68(5): 1233-44, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26636548

RESUMEN

OBJECTIVE: Type I interferon (IFN) signaling is a central pathogenic pathway in systemic lupus erythematosus (SLE), and therapeutics targeting type I IFN signaling are in development. Multiple proteins with overlapping functions play a role in IFN signaling, but the signaling events downstream of receptor engagement are unclear. This study was undertaken to investigate the roles of the type I and type II IFN signaling components IFN-α/ß/ω receptor 2 (IFNAR-2), IFN regulatory factor 9 (IRF-9), and STAT-1 in a mouse model of SLE. METHODS: We used immunohistochemical staining and highly multiplexed assays to characterize pathologic changes in histology, autoantibody production, cytokine/chemokine profiles, and STAT phosphorylation in order to investigate the individual roles of IFNAR-2, IRF-9, and STAT-1 in MRL/lpr mice. RESULTS: We found that STAT-1(-/-) mice, but not IRF-9(-/-) or IFNAR-2(-/-) mice, developed interstitial nephritis characterized by infiltration with retinoic acid receptor-related orphan nuclear receptor γt-positive lymphocytes, macrophages, and eosinophils. Despite pronounced interstitial kidney disease and abnormal kidney function, STAT-1(-/-) mice had decreased proteinuria, glomerulonephritis, and autoantibody production. Phosphospecific flow cytometry revealed shunting of STAT phosphorylation from STAT-1 to STAT-3/4. CONCLUSION: We describe unique contributions of STAT-1 to pathology in different kidney compartments in a mouse model, and provide potentially novel insight into tubulointerstitial nephritis, a poorly understood complication that predicts end-stage kidney disease in SLE patients.


Asunto(s)
Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Lupus Eritematoso Sistémico/genética , Nefritis Intersticial/genética , Receptor de Interferón alfa y beta/genética , Factor de Transcripción STAT1/genética , Células Th17/inmunología , Animales , Formación de Anticuerpos , Autoanticuerpos/inmunología , Técnica del Anticuerpo Fluorescente , Glomerulonefritis/genética , Glomerulonefritis/inmunología , Interferón Tipo I , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/inmunología , Interferón gamma , Riñón/patología , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos MRL lpr , Ratones Noqueados , Nefritis Intersticial/inmunología , Nefritis Intersticial/patología , Proteinuria/genética , Proteinuria/inmunología , Receptor de Interferón alfa y beta/inmunología , Factor de Transcripción STAT1/inmunología
10.
J Clin Invest ; 123(6): 2447-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23728179

RESUMEN

Activation of TLR9 by direct injection of unmethylated CpG nucleotides into a tumor can induce a therapeutic immune response; however, Tregs eventually inhibit the antitumor immune response and thereby limit the power of cancer immunotherapies. In tumor-bearing mice, we found that Tregs within the tumor preferentially express the cell surface markers CTLA-4 and OX40. We show that intratumoral coinjection of anti-CTLA-4 and anti-OX40 together with CpG depleted tumor-infiltrating Tregs. This in situ immunomodulation, which was performed with low doses of antibodies in a single tumor, generated a systemic antitumor immune response that eradicated disseminated disease in mice. Further, this treatment modality was effective against established CNS lymphoma with leptomeningeal metastases, sites that are usually considered to be tumor cell sanctuaries in the context of conventional systemic therapy. These results demonstrate that antitumor immune effectors elicited by local immunomodulation can eradicate tumor cells at distant sites. We propose that, rather than using mAbs to target cancer cells systemically, mAbs could be used to target the tumor infiltrative immune cells locally, thereby eliciting a systemic immune response.


Asunto(s)
Neoplasias Encefálicas/terapia , Linfoma/terapia , Neoplasias Meníngeas/terapia , Linfocitos T Reguladores/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Antígeno CTLA-4/inmunología , Antígeno CTLA-4/metabolismo , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Factores Inmunológicos/administración & dosificación , Inmunomodulación , Inmunoterapia , Inyecciones Intralesiones , Depleción Linfocítica , Linfoma/inmunología , Linfoma/patología , Neoplasias Meníngeas/inmunología , Neoplasias Meníngeas/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/farmacología , Receptores OX40/inmunología , Linfocitos T Reguladores/metabolismo , Receptor Toll-Like 9/agonistas , Carga Tumoral
11.
Nat Neurosci ; 14(9): 1142-9, 2011 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-21804537

RESUMEN

In multiple sclerosis and the experimental autoimmune encephalitis (EAE) mouse model, two pools of morphologically indistinguishable phagocytic cells, microglia and inflammatory macrophages, accrue from proliferating resident precursors and recruitment of blood-borne progenitors, respectively. Whether these cell types are functionally equivalent is hotly debated, but is challenging to address experimentally. Using a combination of parabiosis and myeloablation to replace circulating progenitors without affecting CNS-resident microglia, we found a strong correlation between monocyte infiltration and progression to the paralytic stage of EAE. Inhibition of chemokine receptor-dependent recruitment of monocytes to the CNS blocked EAE progression, suggesting that these infiltrating cells are essential for pathogenesis. Finally, we found that, although microglia can enter the cell cycle and return to quiescence following remission, recruited monocytes vanish, and therefore do not ultimately contribute to the resident microglial pool. In conclusion, we identified two distinct subsets of myelomonocytic cells with distinct roles in neuroinflammation and disease progression.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Microglía/fisiología , Monocitos/fisiología , Animales , Trasplante de Médula Ósea/métodos , Bromodesoxiuridina/metabolismo , Antígeno CD11b/metabolismo , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/prevención & control , Encefalomielitis Autoinmune Experimental/cirugía , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Inflamación/terapia , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Agonistas Mieloablativos/efectos adversos , Parabiosis/métodos , Receptores CCR2/deficiencia , Receptores de Interleucina-8A/genética , Índice de Severidad de la Enfermedad , Factores de Tiempo , Irradiación Corporal Total/efectos adversos
12.
Nat Neurosci ; 10(12): 1538-43, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18026097

RESUMEN

Microgliosis is a common response to multiple types of damage in the CNS. However, the origin of the cells involved in this process is still controversial and the relative importance of local expansion versus recruitment of microglia progenitors from the bloodstream is unclear. Here, we investigated the origin of microglia using chimeric animals obtained by parabiosis. We found no evidence of microglia progenitor recruitment from the circulation in denervation or CNS neurodegenerative disease, suggesting that maintenance and local expansion of microglia are solely dependent on the self-renewal of CNS resident cells in these models.


Asunto(s)
Enfermedades del Sistema Nervioso Central/patología , Sistema Nervioso Central/patología , Microglía/fisiología , Degeneración Nerviosa/patología , Animales , Axotomía/métodos , Trasplante de Médula Ósea/métodos , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Enfermedades del Sistema Nervioso Central/complicaciones , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/cirugía , Modelos Animales de Enfermedad , Enfermedades del Nervio Facial/patología , Enfermedades del Nervio Facial/fisiopatología , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos , Degeneración Nerviosa/etiología , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/cirugía , Parabiosis/métodos , Quimera por Radiación , Células Madre/fisiología , Superóxido Dismutasa/genética , Factores de Tiempo
13.
Glia ; 53(7): 744-53, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16518833

RESUMEN

Amyotrophic lateral sclerosis (ALS) is associated with increased numbers of microglia within the central nervous system (CNS). However, it is unknown whether the microgliosis results from proliferation of CNS resident microglia, or recruitment of bone marrow (BM)-derived microglial precursors. Here we assess the distribution and number of BM-derived cells in spinal cord using transplantation of green fluorescent protein (GFP)-labeled BM cells into myelo-ablated mice over-expressing human mutant superoxide dismutase 1 (mSOD), a murine model of ALS. Transplantation of GFP+ BM did not affect the rate of disease progression in mSOD mice. Mean numbers of microglia and GFP+ cells in spinal cords of control mice were not significantly different from those in asymptomatic mSOD mice and showed no change with animal age. The number of GFP+ cells and microglia (F4/80+ and CD11b+ cells) within the spinal cord of mSOD mice increased compared to age-matched controls at a time when mSOD mice exhibited disease symptoms, continuing up to disease end-stage. Although we observed an increase in the number of GFP+ cells in spinal cords of mSOD mice with disease symptoms, mean numbers of GFP+ F4/80+ cells comprised less than 20% of all F4/80+ cells and did not increase with disease progression. Furthermore, the relative rates of proliferation in CD45+GFP- and CD45+GFP+ cells were comparable. Thus, we demonstrate that the microgliosis present in spinal cord tissue of mSOD mice is primarily due to an expansion of resident microglia and not to the recruitment of microglial precursors from the circulation.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Células de la Médula Ósea/citología , Linaje de la Célula/fisiología , Gliosis/fisiopatología , Microglía/citología , Médula Espinal/fisiopatología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Recuento de Células , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Gliosis/metabolismo , Gliosis/patología , Proteínas Fluorescentes Verdes , Humanos , Antígenos Comunes de Leucocito/inmunología , Ratones , Ratones Transgénicos , Microglía/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...