Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Acta Neuropathol ; 147(1): 78, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38695952

RESUMEN

Aging is associated with cell senescence and is the major risk factor for AD. We characterized premature cell senescence in postmortem brains from non-diseased controls (NDC) and donors with Alzheimer's disease (AD) using imaging mass cytometry (IMC) and single nuclear RNA (snRNA) sequencing (> 200,000 nuclei). We found increases in numbers of glia immunostaining for galactosidase beta (> fourfold) and p16INK4A (up to twofold) with AD relative to NDC. Increased glial expression of genes related to senescence was associated with greater ß-amyloid load. Prematurely senescent microglia downregulated phagocytic pathways suggesting reduced capacity for ß-amyloid clearance. Gene set enrichment and pseudo-time trajectories described extensive DNA double-strand breaks (DSBs), mitochondrial dysfunction and ER stress associated with increased ß-amyloid leading to premature senescence in microglia. We replicated these observations with independent AD snRNA-seq datasets. Our results describe a burden of senescent glia with AD that is sufficiently high to contribute to disease progression. These findings support the hypothesis that microglia are a primary target for senolytic treatments in AD.


Asunto(s)
Enfermedad de Alzheimer , Senescencia Celular , Transcriptoma , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Humanos , Senescencia Celular/fisiología , Senescencia Celular/genética , Anciano , Masculino , Anciano de 80 o más Años , Femenino , Microglía/patología , Microglía/metabolismo , Encéfalo/patología , Encéfalo/metabolismo , Péptidos beta-Amiloides/metabolismo , Neuroglía/patología , Neuroglía/metabolismo
2.
Glia ; 72(7): 1340-1355, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38597386

RESUMEN

Several in vivo studies have shown that systemic inflammation, mimicked by LPS, triggers an inflammatory response in the CNS, driven by microglia, characterized by an increase in inflammatory cytokines and associated sickness behavior. However, most studies induce relatively high systemic inflammation, not directly compared with the more common low-grade inflammatory events experienced in humans during the life course. Using mice, we investigated the effects of low-grade systemic inflammation during an otherwise healthy early life, and how this may precondition the onset and severity of Alzheimer's disease (AD)-like pathology. Our results indicate that low-grade systemic inflammation induces sub-threshold brain inflammation and promotes microglial proliferation driven by the CSF1R pathway, contrary to the effects caused by high systemic inflammation. In addition, repeated systemic challenges with low-grade LPS induce disease-associated microglia. Finally, using an inducible model of AD-like pathology (Line 102 mice), we observed that preconditioning with repeated doses of low-grade systemic inflammation, prior to APP induction, promotes a detrimental effect later in life, leading to an increase in Aß accumulation and disease-associated microglia. These results support the notion that episodic low-grade systemic inflammation has the potential to influence the onset and severity of age-related neurological disorders, such as AD.


Asunto(s)
Enfermedad de Alzheimer , Inflamación , Lipopolisacáridos , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía , Animales , Microglía/metabolismo , Microglía/patología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Inflamación/patología , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Ratones , Modelos Animales de Enfermedad , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Masculino , Femenino , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Encéfalo/patología , Encéfalo/metabolismo , Péptidos beta-Amiloides/metabolismo , Citocinas/metabolismo
3.
Nat Rev Neurosci ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658739

RESUMEN

Microglia constitute the largest population of parenchymal macrophages in the brain and are considered a unique subset of central nervous system glial cells owing to their extra-embryonic origins in the yolk sac. During development, microglial progenitors readily proliferate and eventually colonize the entire brain. In this Review, we highlight the origins of microglial progenitors and their entry routes into the brain and discuss the various molecular and non-molecular determinants of their fate, which may inform their specific functions. Specifically, we explore recently identified mechanisms that regulate microglial colonization of the brain, including the availability of space, and describe how the expansion of highly proliferative microglial progenitors facilitates the occupation of the microglial niche. Finally, we shed light on the factors involved in establishing microglial identity in the brain.

4.
Glia ; 72(2): 375-395, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37909242

RESUMEN

White matter abnormalities, related to poor cerebral perfusion, are a core feature of small vessel cerebrovascular disease, and critical determinants of vascular cognitive impairment and dementia. Despite this importance there is a lack of treatment options. Proliferation of microglia producing an expanded, reactive population and associated neuroinflammatory alterations have been implicated in the onset and progression of cerebrovascular white matter disease, in patients and in animal models, suggesting that targeting microglial proliferation may exert protection. Colony-stimulating factor-1 receptor (CSF1R) is a key regulator of microglial proliferation. We found that the expression of CSF1R/Csf1r and other markers indicative of increased microglial abundance are significantly elevated in damaged white matter in human cerebrovascular disease and in a clinically relevant mouse model of chronic cerebral hypoperfusion and vascular cognitive impairment. Using the mouse model, we investigated long-term pharmacological CSF1R inhibition, via GW2580, and demonstrated that the expansion of microglial numbers in chronic hypoperfused white matter is prevented. Transcriptomic analysis of hypoperfused white matter tissue showed enrichment of microglial and inflammatory gene sets, including phagocytic genes that were the predominant expression modules modified by CSF1R inhibition. Further, CSF1R inhibition attenuated hypoperfusion-induced white matter pathology and rescued spatial learning impairments and to a lesser extent cognitive flexibility. Overall, this work suggests that inhibition of CSF1R and microglial proliferation mediates protection against chronic cerebrovascular white matter pathology and cognitive deficits. Our study nominates CSF1R as a target for the treatment of vascular cognitive disorders with broader implications for treatment of other chronic white matter diseases.


Asunto(s)
Trastornos Cerebrovasculares , Trastornos del Conocimiento , Disfunción Cognitiva , Leucoencefalopatías , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Sustancia Blanca , Animales , Ratones , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/patología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Receptores del Factor Estimulante de Colonias/metabolismo , Sustancia Blanca/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
5.
Cell Rep ; 42(5): 112425, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37099424

RESUMEN

Microglia arise from the yolk sac and enter the brain during early embryogenesis. Upon entry, microglia undergo in situ proliferation and eventually colonize the entire brain by the third postnatal week in mice. However, the intricacies of their developmental expansion remain unclear. Here, we characterize the proliferative dynamics of microglia during embryonic and postnatal development using complementary fate-mapping techniques. We demonstrate that the developmental colonization of the brain is facilitated by clonal expansion of highly proliferative microglial progenitors that occupy spatial niches throughout the brain. Moreover, the spatial distribution of microglia switches from a clustered to a random pattern between embryonic and late postnatal development. Interestingly, the developmental increase in microglial numbers follows the proportional growth of the brain in an allometric manner until a mosaic distribution has been established. Overall, our findings offer insight into how the competition for space may drive microglial colonization by clonal expansion during development.


Asunto(s)
Encéfalo , Microglía , Ratones , Animales , Saco Vitelino , Desarrollo Embrionario
6.
J Neuroinflammation ; 20(1): 92, 2023 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-37032328

RESUMEN

Neuroinflammation is an important component of many neurodegenerative diseases, whether as a primary cause or a secondary outcome. For that reason, either as diagnostic tools or to monitor progression and/or pharmacological interventions, there is a need for robust biomarkers of neuroinflammation in the brain. Mitochondrial TSPO (18 kDa Translocator protein) is one of few available biomarkers of neuroinflammation for which there are clinically available PET imaging agents. In this study, we further characterised neuroinflammation in a mouse model of prion-induced chronic neurodegeneration (ME7) including a pharmacological intervention via a CSF1R inhibitor. This was achieved by autoradiographic binding of the second-generation TSPO tracer, [3H]PBR28, along with a more comprehensive examination of the cellular contributors to the TSPO signal changes by immunohistochemistry. We observed regional increases of TSPO in the ME7 mouse brains, particularly in the hippocampus, cortex and thalamus. This increased TSPO signal was detected in the cells of microglia/macrophage lineage as well as in astrocytes, endothelial cells and neurons. Importantly, we show that the selective CSF1R inhibitor, JNJ-40346527 (JNJ527), attenuated the disease-dependent increase in TSPO signal, particularly in the dentate gyrus of the hippocampus, where JNJ527 attenuated the number of Iba1+ microglia and neurons, but not GFAP+ astrocytes or endothelial cells. These findings suggest that [3H]PBR28 quantitative autoradiography in combination with immunohistochemistry are important translational tools for detecting and quantifying neuroinflammation, and its treatments, in neurodegenerative disease. Furthermore, we demonstrate that although TSPO overexpression in the ME7 brains was driven by various cell types, the therapeutic effect of the CSF1R inhibitor was primarily to modulate TSPO expression in microglia and neurons, which identifies an important route of biological action of this particular CSF1R inhibitor and provides an example of a cell-specific effect of this type of therapeutic agent on the neuroinflammatory process.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedades por Prión , Ratones , Animales , Microglía/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neuroinflamatorias , Células Endoteliales/metabolismo , Receptores de GABA/metabolismo , Tomografía de Emisión de Positrones/métodos , Macrófagos/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Neuronas/metabolismo , Enfermedades por Prión/metabolismo , Biomarcadores/metabolismo
7.
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
8.
Dev Cell ; 57(17): 2127-2139.e6, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-35977545

RESUMEN

Microglia, the brain's resident macrophages, shape neural development and are key neuroimmune hubs in the pathological signatures of neurodevelopmental disorders. Despite the importance of microglia, their development has not been carefully examined in the human brain, and most of our knowledge derives from rodents. We aimed to address this gap in knowledge by establishing an extensive collection of 97 post-mortem tissues in order to enable quantitative, sex-matched, detailed analysis of microglia across the human lifespan. We identify the dynamics of these cells in the human telencephalon, describing waves in microglial density across gestation, infancy, and childhood, controlled by a balance of proliferation and apoptosis, which track key neurodevelopmental milestones. These profound changes in microglia are also observed in bulk RNA-seq and single-cell RNA-seq datasets. This study provides a detailed insight into the spatiotemporal dynamics of microglia across the human lifespan and serves as a foundation for elucidating how microglia contribute to shaping neurodevelopment in humans.


Asunto(s)
Longevidad , Microglía , Encéfalo/patología , Niño , Humanos , Macrófagos , Neurogénesis
9.
Cell Rep ; 35(10): 109228, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34107254

RESUMEN

The sustained proliferation of microglia is a key hallmark of Alzheimer's disease (AD), accelerating its progression. Here, we aim to understand the long-term impact of the early and prolonged microglial proliferation observed in AD, hypothesizing that extensive and repeated cycling would engender a distinct transcriptional and phenotypic trajectory. We show that the early and sustained microglial proliferation seen in an AD-like model promotes replicative senescence, characterized by increased ßgal activity, a senescence-associated transcriptional signature, and telomere shortening, correlating with the appearance of disease-associated microglia (DAM) and senescent microglial profiles in human post-mortem AD cases. The prevention of early microglial proliferation hinders the development of senescence and DAM, impairing the accumulation of Aß, as well as associated neuritic and synaptic damage. Overall, our results indicate that excessive microglial proliferation leads to the generation of senescent DAM, which contributes to early Aß pathology in AD.


Asunto(s)
Péptidos beta-Amiloides/genética , Senescencia Celular/genética , Microglía/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos
10.
J Neurosci Res ; 99(9): 2216-2227, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34051113

RESUMEN

Oligodendrocyte progenitor cells (OPCs) are responsible for generating oligodendrocytes, the myelinating cells of the CNS. Life-long myelination is promoted by neuronal activity and is essential for neural network plasticity and learning. OPCs are known to contact synapses and it is proposed that neuronal synaptic activity in turn regulates their behavior. To examine this in the adult, we performed unilateral injection of the synaptic blocker botulinum neurotoxin A (BoNT/A) into the hippocampus of adult mice. We confirm BoNT/A cleaves SNAP-25 in the CA1 are of the hippocampus, which has been proven to block neurotransmission. Notably, BoNT/A significantly decreased OPC density and caused their shrinkage, as determined by immunolabeling for the OPC marker NG2. Furthermore, BoNT/A resulted in an overall decrease in the number of OPC processes, as well as a decrease in their lengths and branching frequency. These data indicate that synaptic activity is important for maintaining adult OPC numbers and cellular integrity, which is relevant to pathophysiological scenarios characterized by dysregulation of synaptic activity, such as age-related cognitive decline, Multiple Sclerosis and Alzheimer's disease.


Asunto(s)
Toxinas Botulínicas Tipo A/administración & dosificación , Hipocampo/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Sinapsis/efectos de los fármacos , Animales , Recuento de Células/métodos , Hipocampo/citología , Hipocampo/patología , Inyecciones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Células Precursoras de Oligodendrocitos/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Sinapsis/patología , Sinapsis/fisiología
11.
J Neuroinflammation ; 18(1): 81, 2021 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-33757565

RESUMEN

BACKGROUND: Ischemia can induce rapid activation of microglia in the brain. As key immunocompetent cells, reactive microglia play an important role in pathological development of ischemic stroke. However, the role of activated microglia during the development of ischemia remains controversial. Thus, we aimed to investigate the function of reactive microglia in the early stage of ischemic stroke. METHODS: A Rose Bengal photothrombosis model was applied to induce targeted ischemic stroke in mice. CX3CR1CreER:R26iDTR mice were used to specifically deplete resident microglia through intragastric administration of tamoxifen (Ta) and intraperitoneal injection of diphtheria toxin (DT). At day 3 after ischemic stroke, behavioral tests were performed. After that, mouse brains were collected for further histological analysis and detection of mRNA expression of inflammatory factors. RESULTS: The results showed that specific depletion of microglia resulted in a significant decrease in ischemic infarct volume and improved performance in motor ability 3 days after stroke. Microglial depletion caused a remarkable reduction in the densities of degenerating neurons and inducible nitric oxide synthase positive (iNOS+) cells. Importantly, depleting microglia induced a significant increase in the mRNA expression level of anti-inflammatory factors TGF-ß1, Arg1, IL-10, IL-4, and Ym1 as well as a significant decline of pro-inflammatory factors TNF-α, iNOS, and IL-1ß 3 days after stroke. CONCLUSIONS: These results suggest that activated microglia is an important modulator of the brain's inflammatory response in stroke, contributing to neurological deficit and infarct expansion. Modulation of the inflammatory response through the elimination of microglia at a precise time point may be a promising therapeutic approach for the treatment of cerebral ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Isquemia Encefálica/patología , Gliosis/metabolismo , Gliosis/patología , Gliosis/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Ratones , Ratones Transgénicos , Microglía/patología , Accidente Cerebrovascular/patología
12.
Neuroscientist ; 27(1): 10-29, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32441222

RESUMEN

Autism spectrum disorders (ASDs) are a heterogeneous group of neurodevelopmental disorders of genetic and environmental etiologies. Some ASD cases are syndromic: associated with clinically defined patterns of somatic abnormalities and a neurobehavioral phenotype (e.g., Fragile X syndrome). Many cases, however, are idiopathic or non-syndromic. Such disorders present themselves during the early postnatal period when language, speech, and personality start to develop. ASDs manifest by deficits in social communication and interaction, restricted and repetitive patterns of behavior across multiple contexts, sensory abnormalities across multiple modalities and comorbidities, such as epilepsy among many others. ASDs are disorders of connectivity, as synaptic dysfunction is common to both syndromic and idiopathic forms. While multiple theories have been proposed, particularly in idiopathic ASDs, none address why certain brain areas (e.g., frontotemporal) appear more vulnerable than others or identify factors that may affect phenotypic specificity. In this hypothesis article, we identify possible routes leading to, and the consequences of, altered connectivity and review the evidence of central and peripheral synaptic dysfunction in ASDs. We postulate that phenotypic specificity could arise from aberrant experience-dependent plasticity mechanisms in frontal brain areas and peripheral sensory networks and propose why the vulnerability of these areas could be part of a model to unify preexisting pathophysiological theories.


Asunto(s)
Trastorno del Espectro Autista , Red Nerviosa , Plasticidad Neuronal , Sistema Nervioso Periférico , Corteza Prefrontal , Animales , Trastorno del Espectro Autista/etiología , Trastorno del Espectro Autista/inmunología , Trastorno del Espectro Autista/fisiopatología , Humanos , Red Nerviosa/crecimiento & desarrollo , Red Nerviosa/fisiopatología , Plasticidad Neuronal/fisiología , Sistema Nervioso Periférico/crecimiento & desarrollo , Sistema Nervioso Periférico/fisiopatología , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/fisiopatología
13.
Front Cell Neurosci ; 14: 575082, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33343301

RESUMEN

Myelin disruption is a feature of natural aging and Alzheimer's disease (AD). In the CNS, myelin is produced by oligodendrocytes, which are generated throughout life by oligodendrocyte progenitor cells (OPCs). Here, we examined age-related changes in OPCs in APP/PS1 mice, a model for AD-like pathology, compared with non-transgenic (Tg) age-matched controls. The analysis was performed in the CA1 area of the hippocampus following immunolabeling for NG2 with the nuclear dye Hoescht, to identify OPC and OPC sister cells, a measure of OPC replication. The results indicate a significant decrease in the number of OPCs at 9 months in APP/PS1 mice, compared to age-matched controls, without further decline at 14 months. Also, the number of OPC sister cells declined significantly at 14 months in APP/PS1 mice, which was not observed in age-matched controls. Notably, OPCs also displayed marked morphological changes at 14 months in APP/PS1 mice, characterized by an overall shrinkage of OPC process domains and increased process branching. The results indicate that OPC disruption is a pathological sign in the APP/PS1 mouse model of AD.

14.
Front Immunol ; 11: 579000, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33162994

RESUMEN

The proliferation and activation of microglia, the resident macrophages in the brain, is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and prion disease. Colony stimulating factor 1 receptor (CSF1R) is critically involved in regulating microglial proliferation, and CSF1R blocking strategies have been recently used to modulate microglia in neurodegenerative diseases. However, CSF1R is broadly expressed by many cell types and the impact of its inhibition on the innate immune system is still unclear. CSF1R can be activated by two independent ligands, CSF-1 and interleukin 34 (IL-34). Recently, it has been reported that microglia development and maintenance depend on IL-34 signaling. In this study, we evaluate the inhibition of IL-34 as a novel strategy to reduce microglial proliferation in the ME7 model of prion disease. Selective inhibition of IL-34 showed no effects on peripheral macrophage populations in healthy mice, avoiding the side effects observed after CSF1R inhibition on the systemic compartment. However, we observed a reduction in microglial proliferation after IL-34 inhibition in prion-diseased mice, indicating that microglia could be more specifically targeted by reducing IL-34. Overall, our results highlight the challenges of targeting the CSF1R/IL34 axis in the systemic and central compartments, important for framing any therapeutic effort to tackle microglia/macrophage numbers during brain disease.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Encéfalo/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Interleucinas/antagonistas & inhibidores , Microglía/efectos de los fármacos , Degeneración Nerviosa , Enfermedades por Prión/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/toxicidad , Anticuerpos Neutralizantes/toxicidad , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Genes fms , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interleucinas/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Transducción de Señal
15.
Genes (Basel) ; 11(10)2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-33007925

RESUMEN

In this study, we seek to exclude other pathophysiological mechanisms by which Frmd7 knock-down may cause Idiopathic Infantile Nystagmus (IIN) using the Frmd7.tm1a and Frmd7.tm1b murine models. We used a combination of genetic, histological and visual function techniques to characterize the role of Frmd7 gene in IIN using a novel murine model for the disease. We demonstrate that the Frmd7.tm1b allele represents a more robust model of Frmd7 knock-out at the mRNA level. The expression of Frmd7 was investigated using both antibody staining and X-gal staining confirming previous reports that Frmd7 expression in the retina is restricted to starburst amacrine cells and demonstrating that X-gal staining recapitulates the expression pattern in this model. Thus, it offers a useful tool for further expression studies. We also show that gross retinal morphology and electrophysiology are unchanged in these Frmd7 mutant models when compared with wild-type mice. High-speed eye-tracking recordings of Frmd7 mutant mice confirm a specific horizontal optokinetic reflex defect. In summary, our study confirms the likely role for Frmd7 in the optokinetic reflex in mice mediated by starburst amacrine cells. We show that the Frmd7.tm1b model provides a more robust knock-out than the Frmd7.tm1a model at the mRNA level, although the functional consequence is unchanged. Finally, we establish a robust eye-tracking technique in mice that can be used in a variety of future studies using this model and others. Although our data highlight a deficit in the optiokinetic reflex as a result of the starburst amacrine cells in the retina, this does not rule out the involvement of other cells, in the brain or the retina where Frmd7 is expressed, in the pathophysiology of IIN.


Asunto(s)
Proteínas del Citoesqueleto/genética , Modelos Animales de Enfermedad , Enfermedades Genéticas Ligadas al Cromosoma X , Nistagmo Congénito , Alelos , Células Amacrinas/metabolismo , Animales , Proteínas del Citoesqueleto/metabolismo , Electrorretinografía , Femenino , Expresión Génica , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Masculino , Ratones , Ratones Noqueados , Mutación , Nistagmo Congénito/genética , Nistagmo Congénito/patología , Nistagmo Congénito/fisiopatología , Nistagmo Optoquinético , Retina/metabolismo , Retina/patología , Retina/fisiopatología , Tomografía de Coherencia Óptica
16.
Neurobiol Aging ; 94: 130-139, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32619874

RESUMEN

There is increasing evidence that myelin disruption is related to cognitive decline in Alzheimer's disease (AD). In the CNS, myelin is produced by oligodendrocytes, which are generated throughout life by adult oligodendrocyte progenitor cells (OPCs), also known as NG2-glia. To address whether alterations in myelination are related to age-dependent changes in OPCs, we analyzed NG2 and myelin basic protein (MBP) immunolabelling in the hippocampus of 3×Tg-AD mice at 6 and 24 months of age, compared with non-Tg age-matched controls. There was an age-related decrease in MBP immunostaining and OPC density, together with a decline in the number of OPC sister cells, a measure of OPC replication. Notably, the loss of myelin and OPC sister cells occurred earlier at 6 months in 3xTg-AD, suggesting accelerated aging, although there was not a concomitant decline in OPC numbers at this age, suggesting the observed changes in myelin were not a consequence of replicative exhaustion, but possibly of OPC disruption or senescence. In line with this, a key finding is that compared to age-match controls, OPC displayed marked morphological atrophy at 6 months in 3xTg-AD followed by morphological hypertrophy at 24 months, as deduced from significant changes in total cell surface area, total cell volume, somata volume and branching of main processes. Moreover, we show that hypertrophic OPCs surround and infiltrate amyloid-ß (Aß) plaques, a key pathological hallmark of AD. The results indicate that OPCs undergo complex age-related remodeling in the hippocampus of the 3xTg-AD mouse model. We conclude that OPC disruption is an early pathological sign in AD and is a potential factor in accelerated myelin loss and cognitive decline.


Asunto(s)
Enfermedad de Alzheimer/patología , Oligodendroglía/patología , Células Madre/patología , Envejecimiento/patología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Atrofia , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Hipertrofia , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/metabolismo , Vaina de Mielina/patología
17.
Front Cell Neurosci ; 14: 149, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32581720

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia, affecting two-thirds of people with dementia in the world. To date, no disease-modifying treatments are available to stop or delay the progression of AD. This chronic neurodegenerative disease is dominated by a strong innate immune response, whereby microglia plays a central role as the main resident macrophage of the brain. Recent genome-wide association studies (GWASs) have identified single-nucleotide polymorphisms (SNPs) located in microglial genes and associated with a delayed onset of AD, highlighting the important role of these cells on the onset and/or progression of the disease. These findings have increased the interest in targeting microglia-associated neuroinflammation as a potentially disease-modifying therapeutic approach for AD. In this review we provide an overview on the contribution of microglia to the pathophysiology of AD, focusing on the main regulatory pathways controlling microglial population dynamics during the neuroinflammatory response, such as the colony-stimulating factor 1 receptor (CSF1R), its ligands (the colony-stimulating factor 1 and interleukin 34) and the transcription factor PU.1. We also discuss the current therapeutic strategies targeting proliferation to modulate microglia-associated neuroinflammation and their potential impact on peripheral immune cell populations in the short and long-term. Understanding the effects of immunomodulatory approaches on microglia and other immune cell types might be critical for developing specific, effective, and safe therapies for neurodegenerative diseases.

18.
Cereb Cortex ; 30(7): 4246-4256, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32191258

RESUMEN

The molecular processes underlying the aging-related decline in cognitive performance and memory observed in humans are poorly understood. Studies in rodents have shown a decrease in N-methyl-D-aspartate receptors (NMDARs) that contain the GluN2B subunit in aging synapses, and this decrease is correlated with impaired memory functions. However, the age-dependent contribution of GluN2B-containing receptors to synaptic transmission in human cortical synapses has not been previously studied. We investigated the synaptic contribution of GluN2A and GluN2B-containing NMDARs in adult human neurons using fresh nonpathological temporal cortical tissue resected during neurosurgical procedures. The tissue we obtained fulfilled quality criteria by the absence of inflammation markers and proteomic degradation. We show an age-dependent decline in the NMDA/AMPA receptor ratio in adult human temporal cortical synapses. We demonstrate that GluN2B-containing NMDA receptors contribute to synaptic responses in the adult human brain with a reduced contribution in older individuals. With previous evidence demonstrating the critical role of synaptic GluN2B in regulating synaptic strength and memory storage in mice, this progressive reduction of GluN2B in the human brain during aging may underlie a molecular mechanism in the age-related decline in cognitive abilities and memory observed in humans.


Asunto(s)
Envejecimiento/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Lóbulo Temporal/metabolismo , Adulto , Anciano , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores AMPA/metabolismo , Lóbulo Temporal/citología , Adulto Joven
19.
J Neuroinflammation ; 17(1): 88, 2020 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-32192522

RESUMEN

BACKGROUND: The participation of microglia in CNS development and homeostasis indicate that these cells are pivotal for the regeneration that occurs after demyelination. The clearance of myelin debris and the inflammatory-dependent activation of local oligodendrocyte progenitor cells in a demyelinated lesion is dependent on the activation of M2c microglia, which display both phagocytic and healing functions. Emerging interest has been raised about the role of Wnt/ß-catenin signaling in oligodendrogenesis and myelination. Besides, cytokines and growth factors released by microglia can control the survival, proliferation, migration, and differentiation of neural stem cells (NSCs), contributing to remyelination through the oligodendrocyte specification of this adult neurogenic niche. METHODS: TMEV-IDD model was used to study the contribution of dorsal SVZ stem cells to newly born oligodendrocytes in the corpus callosum following demyelination by (i) en-face dorsal SVZ preparations; (ii) immunohistochemistry; and (iii) cellular tracking. By RT-PCR, we analyzed the expression of Wnt proteins in demyelinated and remyelinating corpus callosum. Using in vitro approaches with microglia cultures and embryonic NSCs, we studied the role of purified myelin, Wnt proteins, and polarized microglia-conditioned medium to NSC proliferation and differentiation. One-way ANOVA followed by Bonferroni's post-hoc test, or a Student's t test were used to establish statistical significance. RESULTS: The demyelination caused by TMEV infection is paralleled by an increase in B1 cells and pinwheels in the dorsal SVZ, resulting in the mobilization of SVZ proliferative progenitors and their differentiation into mature oligodendrocytes. Demyelination decreased the gene expression of Wnt5a and Wnt7a, which was restored during remyelination. In vitro approaches show that Wnt3a enhances NSC proliferation, while Wnt7a and myelin debris promotes oligodendrogenesis from NSCs. As phagocytic M2c microglia secrete Wnt 7a, their conditioned media was found to induce Wnt/ß-Catenin signaling in NSCs promoting an oligodendroglial fate. CONCLUSIONS: We define here the contribution of microglia to Wnt production depending on their activation state, with M1 microglia secreting the Wnt5a protein and M2c microglia secreting Wnt7a. Collectively, our data reveal the role of reparative microglia in NSC oligodendrogenesis with the involvement of Wnt7a.


Asunto(s)
Diferenciación Celular/fisiología , Microglía/metabolismo , Neurogénesis/fisiología , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Proteínas Wnt/metabolismo , Animales , Femenino , Ventrículos Laterales/citología , Ratones , Células Precursoras de Oligodendrocitos/citología , Oligodendroglía/citología , Ratas
20.
Brain ; 142(10): 3243-3264, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31504240

RESUMEN

Neuroinflammation and microglial activation are significant processes in Alzheimer's disease pathology. Recent genome-wide association studies have highlighted multiple immune-related genes in association with Alzheimer's disease, and experimental data have demonstrated microglial proliferation as a significant component of the neuropathology. In this study, we tested the efficacy of the selective CSF1R inhibitor JNJ-40346527 (JNJ-527) in the P301S mouse tauopathy model. We first demonstrated the anti-proliferative effects of JNJ-527 on microglia in the ME7 prion model, and its impact on the inflammatory profile, and provided potential CNS biomarkers for clinical investigation with the compound, including pharmacokinetic/pharmacodynamics and efficacy assessment by TSPO autoradiography and CSF proteomics. Then, we showed for the first time that blockade of microglial proliferation and modification of microglial phenotype leads to an attenuation of tau-induced neurodegeneration and results in functional improvement in P301S mice. Overall, this work strongly supports the potential for inhibition of CSF1R as a target for the treatment of Alzheimer's disease and other tau-mediated neurodegenerative diseases.


Asunto(s)
Imidazoles/farmacología , Microglía/efectos de los fármacos , Piridinas/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Enfermedad de Alzheimer/patología , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Imidazoles/metabolismo , Ratones , Ratones Transgénicos , Microglía/fisiología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neurogénesis , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Piridinas/metabolismo , Receptores de GABA/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Tauopatías/tratamiento farmacológico , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...