Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Brain Behav Immun ; 76: 126-138, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30453020

RESUMEN

The cytokine interleukin-1 (IL-1) is a key contributor to neuroinflammation and brain injury, yet mechanisms by which IL-1 triggers neuronal injury remain unknown. Here we induced conditional deletion of IL-1R1 in brain endothelial cells, neurons and blood cells to assess site-specific IL-1 actions in a model of cerebral ischaemia in mice. Tamoxifen treatment of IL-1R1 floxed (fl/fl) mice crossed with mice expressing tamoxifen-inducible Cre-recombinase under the Slco1c1 promoter resulted in brain endothelium-specific deletion of IL-1R1 and a significant decrease in infarct size (29%), blood-brain barrier (BBB) breakdown (53%) and neurological deficit (40%) compared to vehicle-treated or control (IL-1R1fl/fl) mice. Absence of brain endothelial IL-1 signalling improved cerebral blood flow, followed by reduced neutrophil infiltration and vascular activation 24 h after brain injury. Conditional IL-1R1 deletion in neurons using tamoxifen inducible nestin-Cre mice resulted in reduced neuronal injury (25%) and altered microglia-neuron interactions, without affecting cerebral perfusion or vascular activation. Deletion of IL-1R1 specifically in cholinergic neurons reduced infarct size, brain oedema and improved functional outcome. Ubiquitous deletion of IL-1R1 had no effect on brain injury, suggesting beneficial compensatory mechanisms on other cells against the detrimental effects of IL-1 on endothelial cells and neurons. We also show that IL-1R1 signalling deletion in platelets or myeloid cells does not contribute to brain injury after experimental stroke. Thus, brain endothelial and neuronal (cholinergic) IL-1R1 mediate detrimental actions of IL-1 in the brain in ischaemic stroke. Cell-specific targeting of IL-1R1 in the brain could therefore have therapeutic benefits in stroke and other cerebrovascular diseases.


Asunto(s)
Isquemia Encefálica/inmunología , Interleucina-1/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/fisiología , Citocinas/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Inflamación/metabolismo , Interleucina-1/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Transducción de Señal
2.
Acta Neuropathol ; 136(3): 461-482, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30027450

RESUMEN

Neurotropic herpesviruses can establish lifelong infection in humans and contribute to severe diseases including encephalitis and neurodegeneration. However, the mechanisms through which the brain's immune system recognizes and controls viral infections propagating across synaptically linked neuronal circuits have remained unclear. Using a well-established model of alphaherpesvirus infection that reaches the brain exclusively via retrograde transsynaptic spread from the periphery, and in vivo two-photon imaging combined with high resolution microscopy, we show that microglia are recruited to and isolate infected neurons within hours. Selective elimination of microglia results in a marked increase in the spread of infection and egress of viral particles into the brain parenchyma, which are associated with diverse neurological symptoms. Microglia recruitment and clearance of infected cells require cell-autonomous P2Y12 signalling in microglia, triggered by nucleotides released from affected neurons. In turn, we identify microglia as key contributors to monocyte recruitment into the inflamed brain, which process is largely independent of P2Y12. P2Y12-positive microglia are also recruited to infected neurons in the human brain during viral encephalitis and both microglial responses and leukocyte numbers correlate with the severity of infection. Thus, our data identify a key role for microglial P2Y12 in defence against neurotropic viruses, whilst P2Y12-independent actions of microglia may contribute to neuroinflammation by facilitating monocyte recruitment to the sites of infection.


Asunto(s)
Encéfalo/metabolismo , Infecciones por Herpesviridae/metabolismo , Microglía/metabolismo , Monocitos/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Transducción de Señal/fisiología , Animales , Encéfalo/virología , Ratones , Microglía/virología , Neuronas/metabolismo , Neuronas/virología
3.
Ann Neurol ; 75(5): 670-83, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24644058

RESUMEN

OBJECTIVE: Bacterial infection contributes to diverse noninfectious diseases and worsens outcome after stroke. Streptococcus pneumoniae, the most common infection in patients at risk of stroke, is a major cause of prolonged hospitalization and death of stroke patients, but how infection impacts clinical outcome is not known. METHODS: We induced sustained pulmonary infection by a human S. pneumoniae isolate in naive and comorbid rodents to investigate the effect of infection on vascular and inflammatory responses prior to and after cerebral ischemia. RESULTS: S. pneumoniae infection triggered atherogenesis, led to systemic induction of interleukin (IL) 1, and profoundly exacerbated (50-90%) ischemic brain injury in rats and mice, a response that was more severe in combination with old age and atherosclerosis. Systemic blockade of IL-1 with IL-1 receptor antagonist (IL-1Ra) fully reversed infection-induced exacerbation of brain injury and functional impairment caused by cerebral ischemia. We show that infection-induced systemic inflammation mediates its effects via increasing platelet activation and microvascular coagulation in the brain after cerebral ischemia, as confirmed by reduced brain injury in response to blockade of platelet glycoprotein (GP) Ibα. IL-1 and platelet-mediated signals converge on microglia, as both IL-1Ra and GPIbα blockade reversed the production of IL-1α by microglia in response to cerebral ischemia in infected animals. INTERPRETATION: S. pneumoniae infection augments atherosclerosis and exacerbates ischemic brain injury via IL-1 and platelet-mediated systemic inflammation. These mechanisms may contribute to diverse cardio- and cerebrovascular pathologies in humans.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Interleucina-1/efectos adversos , Complejo GPIb-IX de Glicoproteína Plaquetaria/efectos adversos , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/patología , Streptococcus pneumoniae , Animales , Isquemia Encefálica/microbiología , Progresión de la Enfermedad , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Interleucina-1/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/microbiología , Microglía/patología , Activación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/antagonistas & inhibidores , Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Ratas , Ratas Wistar , Infecciones Estreptocócicas/microbiología
4.
Nat Commun ; 13(1): 6715, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36344570

RESUMEN

Neuronal plasticity has been shown to be causally linked to coincidence detection through dendritic spikes (dSpikes). We demonstrate the existence of SPW-R-associated, branch-specific, local dSpikes and their computational role in basal dendrites of hippocampal PV+ interneurons in awake animals. To measure the entire dendritic arbor of long thin dendrites during SPW-Rs, we used fast 3D acousto-optical imaging through an eccentric deep-brain adapter and ipsilateral local field potential recording. The regenerative calcium spike started at variable, NMDA-AMPA-dependent, hot spots and propagated in both direction with a high amplitude beyond a critical distance threshold (~150 µm) involving voltage-gated calcium channels. A supralinear dendritic summation emerged during SPW-R doublets when two successive SPW-R events coincide within a short temporal window (~150 ms), e.g., during more complex association tasks, and generated large dSpikes with an about 2.5-3-fold amplitude increase which propagated down to the soma. Our results suggest that these doublet-associated dSpikes can work as a dendritic-level temporal and spatial coincidence detector during SPW-R-related network computation in awake mice.


Asunto(s)
Interneuronas , Parvalbúminas , Ratones , Animales , Potenciales de Acción/fisiología , Interneuronas/fisiología , Dendritas/fisiología , Neuronas/fisiología , Hipocampo/fisiología , Células Piramidales/fisiología
5.
J Cereb Blood Flow Metab ; 40(1_suppl): S67-S80, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-31987008

RESUMEN

Selective elimination of microglia from the brain was shown to dysregulate neuronal Ca2+ signaling and to reduce the incidence of spreading depolarization (SD) during cerebral ischemia. However, the mechanisms through which microglia interfere with SD remained unexplored. Here, we identify microglia as essential modulators of the induction and evolution of SD in the physiologically intact brain in vivo. Confocal- and super-resolution microscopy revealed that a series of SDs induced rapid morphological changes in microglia, facilitated microglial process recruitment to neurons and increased the density of P2Y12 receptors (P2Y12R) on recruited microglial processes. In line with this, depolarization and hyperpolarization during SD were microglia- and P2Y12R-dependent. An absence of microglia was associated with altered potassium uptake after SD and increased the number of c-fos-positive neurons, independently of P2Y12R. Thus, the presence of microglia is likely to be essential to maintain the electrical elicitation threshold and to support the full evolution of SD, conceivably by interfering with the extracellular potassium homeostasis of the brain through sustaining [K+]e re-uptake mechanisms.


Asunto(s)
Isquemia Encefálica/fisiopatología , Microglía/metabolismo , Potasio/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratones
6.
Science ; 367(6477): 528-537, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31831638

RESUMEN

Microglia are the main immune cells in the brain and have roles in brain homeostasis and neurological diseases. Mechanisms underlying microglia-neuron communication remain elusive. Here, we identified an interaction site between neuronal cell bodies and microglial processes in mouse and human brain. Somatic microglia-neuron junctions have a specialized nanoarchitecture optimized for purinergic signaling. Activity of neuronal mitochondria was linked with microglial junction formation, which was induced rapidly in response to neuronal activation and blocked by inhibition of P2Y12 receptors. Brain injury-induced changes at somatic junctions triggered P2Y12 receptor-dependent microglial neuroprotection, regulating neuronal calcium load and functional connectivity. Thus, microglial processes at these junctions could potentially monitor and protect neuronal functions.


Asunto(s)
Lesiones Encefálicas/inmunología , Encéfalo/inmunología , Uniones Intercelulares/inmunología , Microglía/inmunología , Neuronas/inmunología , Receptores Purinérgicos P2Y12/fisiología , Animales , Encéfalo/ultraestructura , Lesiones Encefálicas/patología , Calcio , Comunicación Celular/inmunología , Células HEK293 , Humanos , Ratones , Mitocondrias/inmunología , Canales de Potasio Shab/genética , Canales de Potasio Shab/fisiología , Transducción de Señal
7.
Mol Imaging Biol ; 20(6): 952-962, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29736562

RESUMEN

PURPOSE: The purpose of this study was to evaluate a set of widely used nuclear medicine imaging agents as possible methods to study the early effects of systemic inflammation on the living brain in a mouse model of sepsis-associated encephalopathy (SAE). The lipopolysaccharide (LPS)-induced murine systemic inflammation model was selected as a model of SAE. PROCEDURES: C57BL/6 mice were used. A multimodal imaging protocol was carried out on each animal 4 h following the intravenous administration of LPS using the following tracers: [99mTc][2,2-dimethyl-3-[(3E)-3-oxidoiminobutan-2-yl]azanidylpropyl]-[(3E)-3-hydroxyiminobutan-2-yl]azanide ([99mTc]HMPAO) and ethyl-7-[125I]iodo-5-methyl-6-oxo-4H-imidazo[1,5-a][1,4]benzodiazepine-3-carboxylate ([125I]iomazenil) to measure brain perfusion and neuronal damage, respectively; 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) to measure cerebral glucose uptake. We assessed microglia activity on another group of mice using 2-[6-chloro-2-(4-[125I]iodophenyl)-imidazo[1,2-a]pyridin-3-yl]-N-ethyl-N-methyl-acetamide ([125I]CLINME). Radiotracer uptakes were measured in different brain regions and correlated. Microglia activity was also assessed using immunohistochemistry. Brain glutathione levels were measured to investigate oxidative stress. RESULTS: Significantly reduced perfusion values and significantly enhanced [18F]FDG and [125I]CLINME uptake was measured in the LPS-treated group. Following perfusion compensation, enhanced [125I]iomazenil uptake was measured in the LPS-treated group's hippocampus and cerebellum. In this group, both [18F]FDG and [125I]iomazenil uptake showed highly negative correlation to perfusion measured with ([99mTc]HMPAO uptake in all brain regions. No significant differences were detected in brain glutathione levels between the groups. The CD45 and P2Y12 double-labeling immunohistochemistry showed widespread microglia activation in the LPS-treated group. CONCLUSIONS: Our results suggest that [125I]CLINME and [99mTc]HMPAO SPECT can be used to detect microglia activation and brain hypoperfusion, respectively, in the early phase (4 h post injection) of systemic inflammation. We suspect that the enhancement of [18F]FDG and [125I]iomazenil uptake in the LPS-treated group does not necessarily reflect neural hypermetabolism and the lack of neuronal damage. They are most likely caused by processes emerging during neuroinflammation, e.g., microglia activation and/or immune cell infiltration.


Asunto(s)
Encéfalo/diagnóstico por imagen , Neuroimagen/métodos , Trazadores Radiactivos , Cintigrafía/métodos , Encefalopatía Asociada a la Sepsis/diagnóstico , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Fluorodesoxiglucosa F18/farmacocinética , Glucosa/metabolismo , Radioisótopos de Yodo/farmacocinética , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Imagen Multimodal/métodos , Medicina Nuclear/métodos , Tomografía de Emisión de Positrones/métodos , Encefalopatía Asociada a la Sepsis/inducido químicamente , Encefalopatía Asociada a la Sepsis/metabolismo , Encefalopatía Asociada a la Sepsis/patología , Exametazima de Tecnecio Tc 99m/farmacocinética , Tomografía Computarizada de Emisión de Fotón Único/métodos
8.
Nat Commun ; 7: 11499, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-27139776

RESUMEN

Microglia are the main immune cells of the brain and contribute to common brain diseases. However, it is unclear how microglia influence neuronal activity and survival in the injured brain in vivo. Here we develop a precisely controlled model of brain injury induced by cerebral ischaemia combined with fast in vivo two-photon calcium imaging and selective microglial manipulation. We show that selective elimination of microglia leads to a striking, 60% increase in infarct size, which is reversed by microglial repopulation. Microglia-mediated protection includes reduction of excitotoxic injury, since an absence of microglia leads to dysregulated neuronal calcium responses, calcium overload and increased neuronal death. Furthermore, the incidence of spreading depolarization (SD) is markedly reduced in the absence of microglia. Thus, microglia are involved in changes in neuronal network activity and SD after brain injury in vivo that could have important implications for common brain diseases.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Microglía/fisiología , Red Nerviosa/fisiopatología , Neuronas/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Isquemia Encefálica/fisiopatología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microscopía de Fluorescencia por Excitación Multifotónica , Neuroprotección/fisiología , Imagen de Lapso de Tiempo/métodos
9.
J Cereb Blood Flow Metab ; 35(12): 1921-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26219594

RESUMEN

Inflammation that develops in the brain and peripheral organs after stroke contributes profoundly to poor outcome of patients. However, mechanisms through which inflammation impacts on brain injury and overall outcome are improperly understood, in part because the earliest inflammatory events after brain injury are not revealed by current imaging tools. Here, we show that single-photon emission computed tomography (NanoSPECT/CT Plus) allows visualization of blood brain barrier (BBB) injury after experimental stroke well before changes can be detected with magnetic resonance imaging (MRI). Early 99mTc-DTPA (diethylene triamine pentaacetic acid) signal changes predict infarct development and systemic inflammation preceding experimental stroke leads to very early perfusion deficits and increased BBB injury within 2 hours after the onset of ischemia. Acute brain injury also leads to peripheral inflammation and immunosuppression, which contribute to poor outcome of stroke patients. The SPECT imaging revealed early (within 2 hours) changes in perfusion, barrier function and inflammation in the lungs and the gut after experimental stroke, with good predictive value for the development of histopathologic changes at later time points. Collectively, visualization of early inflammatory changes after stroke could open new translational research avenues to elucidate the interactions between central and peripheral inflammation and to evaluate in vivo 'multi-system' effects of putative anti-inflammatory treatments.


Asunto(s)
Isquemia Encefálica/diagnóstico por imagen , Inflamación/diagnóstico por imagen , Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/patología , Isquemia Encefálica/patología , Tracto Gastrointestinal/patología , Procesamiento de Imagen Asistido por Computador , Terapia de Inmunosupresión , Infarto de la Arteria Cerebral Media/diagnóstico por imagen , Infarto de la Arteria Cerebral Media/patología , Inflamación/patología , Pulmón/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/psicología , Radiofármacos , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/patología , Pentetato de Tecnecio Tc 99m
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA