Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 38: 597-620, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-32340575

RESUMEN

Neuroimmunology, albeit a relatively established discipline, has recently sparked numerous exciting findings on microglia, the resident macrophages of the central nervous system (CNS). This review addresses meningeal immunity, a less-studied aspect of neuroimmune interactions. The meninges, a triple layer of membranes-the pia mater, arachnoid mater, and dura mater-surround the CNS, encompassing the cerebrospinal fluid produced by the choroid plexus epithelium. Unlike the adjacent brain parenchyma, the meninges contain a wide repertoire of immune cells. These constitute meningeal immunity, which is primarily concerned with immune surveillance of the CNS, and-according to recent evidence-also participates in postinjury CNS recovery, chronic neurodegenerative conditions, and even higher brain function. Meningeal immunity has recently come under the spotlight owing to the characterization of meningeal lymphatic vessels draining the CNS. Here, we review the current state of our understanding of meningeal immunity and its effects on healthy and diseased brains.


Asunto(s)
Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Susceptibilidad a Enfermedades , Homeostasis , Inmunidad , Meninges/fisiología , Animales , Humanos , Vasos Linfáticos/inmunología , Vasos Linfáticos/metabolismo , Neuroinmunomodulación , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
2.
Cell ; 186(3): 464-466, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36693375

RESUMEN

T cells and their derived cytokines have been shown to modulate brain function. In this issue of Cell, Zhu, Yan, and colleagues demonstrate that opioid use impacts the crosstalk between the CNS and the peripheral immune system. Regulatory T cell (Treg)-derived IFN-γ signaling translates into synaptic weakening in the nucleus accumbens (NAc) to impart withdrawal-induced behavioral dysfunction.


Asunto(s)
Núcleo Accumbens , Trastornos Relacionados con Opioides , Transducción de Señal , Núcleo Accumbens/fisiología , Trastornos Relacionados con Opioides/patología , Citocinas
3.
Cell ; 184(4): 1000-1016.e27, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33508229

RESUMEN

Despite the established dogma of central nervous system (CNS) immune privilege, neuroimmune interactions play an active role in diverse neurological disorders. However, the precise mechanisms underlying CNS immune surveillance remain elusive; particularly, the anatomical sites where peripheral adaptive immunity can sample CNS-derived antigens and the cellular and molecular mediators orchestrating this surveillance. Here, we demonstrate that CNS-derived antigens in the cerebrospinal fluid (CSF) accumulate around the dural sinuses, are captured by local antigen-presenting cells, and are presented to patrolling T cells. This surveillance is enabled by endothelial and mural cells forming the sinus stromal niche. T cell recognition of CSF-derived antigens at this site promoted tissue resident phenotypes and effector functions within the dural meninges. These findings highlight the critical role of dural sinuses as a neuroimmune interface, where brain antigens are surveyed under steady-state conditions, and shed light on age-related dysfunction and neuroinflammatory attack in animal models of multiple sclerosis.


Asunto(s)
Senos Craneales/inmunología , Senos Craneales/fisiología , Duramadre/inmunología , Duramadre/fisiología , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos/líquido cefalorraquídeo , Senescencia Celular , Quimiocina CXCL12/farmacología , Duramadre/irrigación sanguínea , Femenino , Homeostasis , Humanos , Inmunidad , Masculino , Ratones Endogámicos C57BL , Fenotipo , Células del Estroma/citología , Linfocitos T/citología
4.
Cell ; 182(2): 270-296, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32707093

RESUMEN

Mammals have two specialized vascular circulatory systems: the blood vasculature and the lymphatic vasculature. The lymphatic vasculature is a unidirectional conduit that returns filtered interstitial arterial fluid and tissue metabolites to the blood circulation. It also plays major roles in immune cell trafficking and lipid absorption. As we discuss in this review, the molecular characterization of lymphatic vascular development and our understanding of this vasculature's role in pathophysiological conditions has greatly improved in recent years, changing conventional views about the roles of the lymphatic vasculature in health and disease. Morphological or functional defects in the lymphatic vasculature have now been uncovered in several pathological conditions. We propose that subtle asymptomatic alterations in lymphatic vascular function could underlie the variability seen in the body's response to a wide range of human diseases.


Asunto(s)
Vasos Linfáticos/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Historia del Siglo XXI , Humanos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Linfangiogénesis , Enfermedades Linfáticas/genética , Enfermedades Linfáticas/historia , Enfermedades Linfáticas/patología , Metástasis Linfática , Vasos Linfáticos/anatomía & histología , Vasos Linfáticos/citología , Neoplasias/metabolismo , Neoplasias/patología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
5.
Immunity ; 57(6): 1394-1412.e8, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38821054

RESUMEN

Recent single-cell RNA sequencing studies have revealed distinct microglial states in development and disease. These include proliferative-region-associated microglia (PAMs) in developing white matter and disease-associated microglia (DAMs) prevalent in various neurodegenerative conditions. PAMs and DAMs share a similar core gene signature. However, the extent of the dynamism and plasticity of these microglial states, as well as their functional significance, remains elusive, partly due to the lack of specific tools. Here, we generated an inducible Cre driver line, Clec7a-CreERT2, that targets PAMs and DAMs in the brain parenchyma. Utilizing this tool, we profiled labeled cells during development and in several disease models, uncovering convergence and context-dependent differences in PAM and DAM gene expression. Through long-term tracking, we demonstrated microglial state plasticity. Lastly, we specifically depleted DAMs in demyelination, revealing their roles in disease recovery. Together, we provide a versatile genetic tool to characterize microglial states in CNS development and disease.


Asunto(s)
Plasticidad de la Célula , Microglía , Remielinización , Microglía/fisiología , Animales , Ratones , Plasticidad de la Célula/genética , Enfermedades Desmielinizantes/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales de Enfermedad , Encéfalo , Vaina de Mielina/metabolismo , Sustancia Blanca/patología
6.
Annu Rev Neurosci ; 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38648267

RESUMEN

Since its recent discovery, the meningeal lymphatic system has reshaped our understanding of central nervous system (CNS) fluid exchange, waste clearance, immune cell trafficking, and immune privilege. Meningeal lymphatics have also been demonstrated to functionally modify the outcome of neurological disorders and their responses to treatment, including brain tumors, inflammatory diseases such as multiple sclerosis, CNS injuries, and neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. In this review, we discuss recent evidence of the contribution of meningeal lymphatics to neurological diseases, as well as the available experimental methods for manipulating meningeal lymphatics in these conditions. Finally, we also provide a discussion of the pressing questions and challenges in utilizing meningeal lymphatics as a prime target for CNS therapeutic intervention and possibly drug delivery for brain disorders.

7.
Nat Immunol ; 21(11): 1421-1429, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32929273

RESUMEN

Interleukin (IL)-17a has been highly conserved during evolution of the vertebrate immune system and widely studied in contexts of infection and autoimmunity. Studies suggest that IL-17a promotes behavioral changes in experimental models of autism and aggregation behavior in worms. Here, through a cellular and molecular characterization of meningeal γδ17 T cells, we defined the nearest central nervous system-associated source of IL-17a under homeostasis. Meningeal γδ T cells express high levels of the chemokine receptor CXCR6 and seed meninges shortly after birth. Physiological release of IL-17a by these cells was correlated with anxiety-like behavior in mice and was partially dependent on T cell receptor engagement and commensal-derived signals. IL-17a receptor was expressed in cortical glutamatergic neurons under steady state and its genetic deletion decreased anxiety-like behavior in mice. Our findings suggest that IL-17a production by meningeal γδ17 T cells represents an evolutionary bridge between this conserved anti-pathogen molecule and survival behavioral traits in vertebrates.


Asunto(s)
Ansiedad/etiología , Ansiedad/metabolismo , Interleucina-17/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Ansiedad/psicología , Conducta Animal , Proliferación Celular , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Duramadre , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Interleucina-17/genética , Meninges/inmunología , Meninges/metabolismo , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Transducción de Señal , Transcriptoma
8.
Cell ; 169(7): 1172-1174, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28622503

RESUMEN

The role of microglia in neurodegenerative diseases has been controversial. In this issue, Keren-Shaul et al. identify a unique population of disease-associated microglia (DAM) that develop in two steps and may help to restrict damage in Alzheimer and related diseases.


Asunto(s)
Microglía , Enfermedades Neurodegenerativas , Humanos
9.
Immunity ; 55(8): 1448-1465.e6, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35931085

RESUMEN

Brain macrophage populations include parenchymal microglia, border-associated macrophages, and recruited monocyte-derived cells; together, they control brain development and homeostasis but are also implicated in aging pathogenesis and neurodegeneration. The phenotypes, localization, and functions of each population in different contexts have yet to be resolved. We generated a murine brain myeloid scRNA-seq integration to systematically delineate brain macrophage populations. We show that the previously identified disease-associated microglia (DAM) population detected in murine Alzheimer's disease models actually comprises two ontogenetically and functionally distinct cell lineages: embryonically derived triggering receptor expressed on myeloid cells 2 (TREM2)-dependent DAM expressing a neuroprotective signature and monocyte-derived TREM2-expressing disease inflammatory macrophages (DIMs) accumulating in the brain during aging. These two distinct populations appear to also be conserved in the human brain. Herein, we generate an ontogeny-resolved model of brain myeloid cell heterogeneity in development, homeostasis, and disease and identify cellular targets for the treatment of neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Envejecimiento , Enfermedad de Alzheimer/genética , Animales , Encéfalo/patología , Humanos , Macrófagos/patología , Glicoproteínas de Membrana , Ratones , Microglía/patología , Receptores Inmunológicos
10.
Nat Immunol ; 24(5): 741-742, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37095376
11.
Nature ; 627(8002): 157-164, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38418877

RESUMEN

The accumulation of metabolic waste is a leading cause of numerous neurological disorders, yet we still have only limited knowledge of how the brain performs self-cleansing. Here we demonstrate that neural networks synchronize individual action potentials to create large-amplitude, rhythmic and self-perpetuating ionic waves in the interstitial fluid of the brain. These waves are a plausible mechanism to explain the correlated potentiation of the glymphatic flow1,2 through the brain parenchyma. Chemogenetic flattening of these high-energy ionic waves largely impeded cerebrospinal fluid infiltration into and clearance of molecules from the brain parenchyma. Notably, synthesized waves generated through transcranial optogenetic stimulation substantially potentiated cerebrospinal fluid-to-interstitial fluid perfusion. Our study demonstrates that neurons serve as master organizers for brain clearance. This fundamental principle introduces a new theoretical framework for the functioning of macroscopic brain waves.


Asunto(s)
Encéfalo , Líquido Cefalorraquídeo , Líquido Extracelular , Neuronas , Potenciales de Acción , Encéfalo/citología , Encéfalo/metabolismo , Ondas Encefálicas/fisiología , Líquido Cefalorraquídeo/metabolismo , Líquido Extracelular/metabolismo , Sistema Glinfático/metabolismo , Cinética , Red Nerviosa/fisiología , Neuronas/metabolismo , Optogenética , Tejido Parenquimatoso/metabolismo , Iones/metabolismo
12.
Nature ; 627(8002): 165-173, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38326613

RESUMEN

The arachnoid barrier delineates the border between the central nervous system and dura mater. Although the arachnoid barrier creates a partition, communication between the central nervous system and the dura mater is crucial for waste clearance and immune surveillance1,2. How the arachnoid barrier balances separation and communication is poorly understood. Here, using transcriptomic data, we developed transgenic mice to examine specific anatomical structures that function as routes across the arachnoid barrier. Bridging veins create discontinuities where they cross the arachnoid barrier, forming structures that we termed arachnoid cuff exit (ACE) points. The openings that ACE points create allow the exchange of fluids and molecules between the subarachnoid space and the dura, enabling the drainage of cerebrospinal fluid and limited entry of molecules from the dura to the subarachnoid space. In healthy human volunteers, magnetic resonance imaging tracers transit along bridging veins in a similar manner to access the subarachnoid space. Notably, in neuroinflammatory conditions such as experimental autoimmune encephalomyelitis, ACE points also enable cellular trafficking, representing a route for immune cells to directly enter the subarachnoid space from the dura mater. Collectively, our results indicate that ACE points are a critical part of the anatomy of neuroimmune communication in both mice and humans that link the central nervous system with the dura and its immunological diversity and waste clearance systems.


Asunto(s)
Aracnoides , Encéfalo , Duramadre , Animales , Humanos , Ratones , Aracnoides/anatomía & histología , Aracnoides/irrigación sanguínea , Aracnoides/inmunología , Aracnoides/metabolismo , Transporte Biológico , Encéfalo/anatomía & histología , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Encéfalo/metabolismo , Duramadre/anatomía & histología , Duramadre/irrigación sanguínea , Duramadre/inmunología , Duramadre/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Perfilación de la Expresión Génica , Imagen por Resonancia Magnética , Ratones Transgénicos , Espacio Subaracnoideo/anatomía & histología , Espacio Subaracnoideo/irrigación sanguínea , Espacio Subaracnoideo/inmunología , Espacio Subaracnoideo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Venas/metabolismo
13.
Nature ; 615(7953): 668-677, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36890231

RESUMEN

Extracellular deposition of amyloid-ß as neuritic plaques and intracellular accumulation of hyperphosphorylated, aggregated tau as neurofibrillary tangles are two of the characteristic hallmarks of Alzheimer's disease1,2. The regional progression of brain atrophy in Alzheimer's disease highly correlates with tau accumulation but not amyloid deposition3-5, and the mechanisms of tau-mediated neurodegeneration remain elusive. Innate immune responses represent a common pathway for the initiation and progression of some neurodegenerative diseases. So far, little is known about the extent or role of the adaptive immune response and its interaction with the innate immune response in the presence of amyloid-ß or tau pathology6. Here we systematically compared the immunological milieux in the brain of mice with amyloid deposition or tau aggregation and neurodegeneration. We found that mice with tauopathy but not those with amyloid deposition developed a unique innate and adaptive immune response and that depletion of microglia or T cells blocked tau-mediated neurodegeneration. Numbers of T cells, especially those of cytotoxic T cells, were markedly increased in areas with tau pathology in mice with tauopathy and in the Alzheimer's disease brain. T cell numbers correlated with the extent of neuronal loss, and the cells dynamically transformed their cellular characteristics from activated to exhausted states along with unique TCR clonal expansion. Inhibition of interferon-γ and PDCD1 signalling both significantly ameliorated brain atrophy. Our results thus reveal a tauopathy- and neurodegeneration-related immune hub involving activated microglia and T cell responses, which could serve as therapeutic targets for preventing neurodegeneration in Alzheimer's disease and primary tauopathies.


Asunto(s)
Encéfalo , Microglía , Ovillos Neurofibrilares , Linfocitos T , Tauopatías , Animales , Ratones , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/metabolismo , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Microglía/inmunología , Microglía/metabolismo , Ovillos Neurofibrilares/inmunología , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Proteínas tau/inmunología , Proteínas tau/metabolismo , Tauopatías/inmunología , Tauopatías/metabolismo , Tauopatías/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Placa Amiloide/inmunología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología , Células Clonales/inmunología , Células Clonales/metabolismo , Células Clonales/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Inmunidad Innata
14.
Nature ; 612(7940): 417-429, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36517712

RESUMEN

The concept of immune privilege suggests that the central nervous system is isolated from the immune system. However, recent studies have highlighted the borders of the central nervous system as central sites of neuro-immune interactions. Although the nervous and immune systems both function to maintain homeostasis, under rare circumstances, they can develop pathological interactions that lead to neurological or psychiatric diseases. Here we discuss recent findings that dissect the key anatomical, cellular and molecular mechanisms that enable neuro-immune responses at the borders of the brain and spinal cord and the implications of these interactions for diseases of the central nervous system.


Asunto(s)
Encéfalo , Sistema Inmunológico , Neuroinmunomodulación , Encéfalo/inmunología , Encéfalo/fisiología , Encéfalo/fisiopatología , Sistema Inmunológico/inmunología , Sistema Inmunológico/fisiología , Sistema Inmunológico/fisiopatología , Neuroinmunomodulación/inmunología , Neuroinmunomodulación/fisiología , Médula Espinal/inmunología , Médula Espinal/fisiología , Médula Espinal/fisiopatología , Humanos , Enfermedades del Sistema Nervioso/inmunología , Enfermedades del Sistema Nervioso/fisiopatología , Enfermedades del Sistema Nervioso/psicología
15.
Nature ; 611(7936): 585-593, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36352225

RESUMEN

Macrophages are important players in the maintenance of tissue homeostasis1. Perivascular and leptomeningeal macrophages reside near the central nervous system (CNS) parenchyma2, and their role in CNS physiology has not been sufficiently well studied. Given their continuous interaction with the cerebrospinal fluid (CSF) and strategic positioning, we refer to these cells collectively as parenchymal border macrophages (PBMs). Here we demonstrate that PBMs regulate CSF flow dynamics. We identify a subpopulation of PBMs that express high levels of CD163 and LYVE1 (scavenger receptor proteins), closely associated with the brain arterial tree, and show that LYVE1+ PBMs regulate arterial motion that drives CSF flow. Pharmacological or genetic depletion of PBMs led to accumulation of extracellular matrix proteins, obstructing CSF access to perivascular spaces and impairing CNS perfusion and clearance. Ageing-associated alterations in PBMs and impairment of CSF dynamics were restored after intracisternal injection of macrophage colony-stimulating factor. Single-nucleus RNA sequencing data obtained from patients with Alzheimer's disease (AD) and from non-AD individuals point to changes in phagocytosis, endocytosis and interferon-γ signalling on PBMs, pathways that are corroborated in a mouse model of AD. Collectively, our results identify PBMs as new cellular regulators of CSF flow dynamics, which could be targeted pharmacologically to alleviate brain clearance deficits associated with ageing and AD.


Asunto(s)
Sistema Nervioso Central , Líquido Cefalorraquídeo , Macrófagos , Tejido Parenquimatoso , Animales , Ratones , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Líquido Cefalorraquídeo/metabolismo , Macrófagos/fisiología , Meninges/citología , Reología , Proteínas de la Matriz Extracelular/metabolismo , Envejecimiento/metabolismo , Fagocitosis , Endocitosis , Interferón gamma/metabolismo , Tejido Parenquimatoso/citología , Humanos
16.
Trends Immunol ; 45(5): 329-337, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600001

RESUMEN

Neurodegenerative disorders present major challenges to global health, exacerbated by an aging population and the absence of therapies. Despite diverse pathological manifestations, they share a common hallmark, loosely termed 'neuroinflammation'. The prevailing dogma is that the immune system is an active contributor to neurodegeneration; however, recent evidence challenges this. By analogy with road construction, which causes temporary closures and disruptions, the immune system's actions in the central nervous system (CNS) might initially appear destructive, and might even cause harm, while aiming to combat neurodegeneration. We propose that the application of cellular immunotherapies to coordinate the immune response towards remodeling might pave the way for new modes of tackling the roadblocks of neurodegenerative diseases.


Asunto(s)
Inmunoterapia , Enfermedades Neurodegenerativas , Animales , Humanos , Sistema Nervioso Central/inmunología , Inmunoterapia/métodos , Enfermedades Neurodegenerativas/terapia , Enfermedades Neurodegenerativas/inmunología
17.
Immunity ; 48(2): 380-395.e6, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29426702

RESUMEN

Individual reports suggest that the central nervous system (CNS) contains multiple immune cell types with diverse roles in tissue homeostasis, immune defense, and neurological diseases. It has been challenging to map leukocytes across the entire brain, and in particular in pathology, where phenotypic changes and influx of blood-derived cells prevent a clear distinction between reactive leukocyte populations. Here, we applied high-dimensional single-cell mass and fluorescence cytometry, in parallel with genetic fate mapping systems, to identify, locate, and characterize multiple distinct immune populations within the mammalian CNS. Using this approach, we revealed that microglia, several subsets of border-associated macrophages and dendritic cells coexist in the CNS at steady state and exhibit disease-specific transformations in the immune microenvironment during aging and in models of Alzheimer's disease and multiple sclerosis. Together, these data and the described framework provide a resource for the study of disease mechanisms, potential biomarkers, and therapeutic targets in CNS disease.


Asunto(s)
Envejecimiento/inmunología , Sistema Nervioso Central/inmunología , Leucocitos/inmunología , Macrófagos/inmunología , Animales , Células Dendríticas/inmunología , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/inmunología , Análisis de la Célula Individual
18.
PLoS Biol ; 22(5): e3002596, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38718086

RESUMEN

Autism spectrum disorders (ASD) frequently accompany macrocephaly, which often involves hydrocephalic enlargement of brain ventricles. Katnal2 is a microtubule-regulatory protein strongly linked to ASD, but it remains unclear whether Katnal2 knockout (KO) in mice leads to microtubule- and ASD-related molecular, synaptic, brain, and behavioral phenotypes. We found that Katnal2-KO mice display ASD-like social communication deficits and age-dependent progressive ventricular enlargements. The latter involves increased length and beating frequency of motile cilia on ependymal cells lining ventricles. Katnal2-KO hippocampal neurons surrounded by enlarged lateral ventricles show progressive synaptic deficits that correlate with ASD-like transcriptomic changes involving synaptic gene down-regulation. Importantly, early postnatal Katnal2 re-expression prevents ciliary, ventricular, and behavioral phenotypes in Katnal2-KO adults, suggesting a causal relationship and a potential treatment. Therefore, Katnal2 negatively regulates ependymal ciliary function and its deletion in mice leads to ependymal ciliary hyperfunction and hydrocephalus accompanying ASD-related behavioral, synaptic, and transcriptomic changes.


Asunto(s)
Trastorno del Espectro Autista , Cilios , Epéndimo , Ratones Noqueados , Fenotipo , Animales , Masculino , Ratones , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/fisiopatología , Conducta Animal , Cilios/metabolismo , Modelos Animales de Enfermedad , Epéndimo/metabolismo , Hipocampo/metabolismo , Hidrocefalia/genética , Hidrocefalia/metabolismo , Hidrocefalia/patología , Hidrocefalia/fisiopatología , Katanina/metabolismo , Katanina/genética , Ratones Endogámicos C57BL , Neuronas/metabolismo , Sinapsis/metabolismo , Transcriptoma/genética
19.
Nature ; 593(7858): 255-260, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33911285

RESUMEN

Alzheimer's disease (AD) is the most prevalent cause of dementia1. Although there is no effective treatment for AD, passive immunotherapy with monoclonal antibodies against amyloid beta (Aß) is a promising therapeutic strategy2,3. Meningeal lymphatic drainage has an important role in the accumulation of Aß in the brain4, but it is not known whether modulation of meningeal lymphatic function can influence the outcome of immunotherapy in AD. Here we show that ablation of meningeal lymphatic vessels in 5xFAD mice (a mouse model of amyloid deposition that expresses five mutations found in familial AD) worsened the outcome of mice treated with anti-Aß passive immunotherapy by exacerbating the deposition of Aß, microgliosis, neurovascular dysfunction, and behavioural deficits. By contrast, therapeutic delivery of vascular endothelial growth factor C improved clearance of Aß by monoclonal antibodies. Notably, there was a substantial overlap between the gene signature of microglia from 5xFAD mice with impaired meningeal lymphatic function and the transcriptional profile of activated microglia from the brains of individuals with AD. Overall, our data demonstrate that impaired meningeal lymphatic drainage exacerbates the microglial inflammatory response in AD and that enhancement of meningeal lymphatic function combined with immunotherapies could lead to better clinical outcomes.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Inmunoterapia , Vasos Linfáticos/inmunología , Meninges/inmunología , Microglía/inmunología , Envejecimiento/efectos de los fármacos , Envejecimiento/inmunología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/efectos de los fármacos , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Encéfalo/irrigación sanguínea , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Modelos Animales de Enfermedad , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Masculino , Meninges/irrigación sanguínea , Meninges/citología , Ratones , Microglía/citología , Microglía/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/farmacología
20.
Immunity ; 46(6): 943-956, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28636961

RESUMEN

The central nervous system (CNS) and its meningeal coverings accommodate a diverse myeloid compartment that includes parenchymal microglia and perivascular macrophages, as well as choroid plexus and meningeal macrophages, dendritic cells, and granulocytes. These myeloid populations enjoy an intimate relationship with the CNS, where they play an essential role in both health and disease. Although the importance of these cells is clearly recognized, their exact function in the CNS continues to be explored. Here, we review the subsets of myeloid cells that inhabit the parenchyma, meninges, and choroid plexus and discuss their roles in CNS homeostasis. We also discuss the role of these cells in various neurological pathologies, such as autoimmunity, mechanical injury, neurodegeneration, and infection. We highlight the neuroprotective nature of certain myeloid cells by emphasizing their therapeutic potential for the treatment of neurological conditions.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Plexo Coroideo/inmunología , Infecciones/inmunología , Células Mieloides/fisiología , Enfermedades Neurodegenerativas/inmunología , Neuroinmunomodulación , Heridas y Lesiones/inmunología , Animales , Sistema Nervioso Central , Humanos , Meninges/inmunología , Neuroprotección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA