Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Cent Sci ; 10(5): 1105-1114, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38799654

RESUMO

Cyclooxygenase-2 (COX-2) is an enzyme that plays a pivotal role in peripheral inflammation and pain via the prostaglandin pathway. In the central nervous system (CNS), COX-2 is implicated in neurodegenerative and psychiatric disorders as a potential therapeutic target and biomarker. However, clinical studies with COX-2 have yielded inconsistent results, partly due to limited mechanistic understanding of how COX-2 activity relates to CNS pathology. Therefore, developing COX-2 positron emission tomography (PET) radiotracers for human neuroimaging is of interest. This study introduces [11C]BRD1158, which is a potent and uniquely fast-binding, selective COX-2 PET radiotracer. [11C]BRD1158 was developed by prioritizing potency at COX-2, isoform selectivity over COX-1, fast binding kinetics, and free fraction in the brain. Evaluated through in vivo PET neuroimaging in rodent models with human COX-2 overexpression, [11C]BRD1158 demonstrated high brain uptake, fast target-engagement, functional reversibility, and excellent specific binding, which is advantageous for human imaging applications. Lastly, post-mortem samples from Huntington's disease (HD) patients and preclinical HD mouse models showed that COX-2 levels were elevated specifically in disease-affected brain regions, primarily from increased expression in microglia. These findings indicate that COX-2 holds promise as a novel clinical marker of HD onset and progression, one of many potential applications of [11C]BRD1158 human PET.

2.
Brain Behav Immun ; 119: 317-332, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38552925

RESUMO

Complement proteins facilitate synaptic elimination during neurodevelopmental pruning, but neural complement regulation is not well understood. CUB and Sushi Multiple Domains 1 (CSMD1) can regulate complement activity in vitro, is expressed in the brain, and is associated with increased schizophrenia risk. Beyond this, little is known about CSMD1 including whether it regulates complement activity in the brain or otherwise plays a role in neurodevelopment. We used biochemical, immunohistochemical, and proteomic techniques to examine the regional, cellular, and subcellular distribution as well as protein interactions of CSMD1 in the brain. To evaluate whether CSMD1 is involved in complement-mediated synapse elimination, we examined Csmd1-knockout mice and CSMD1-knockout human stem cell-derived neurons. We interrogated synapse and circuit development of the mouse visual thalamus, a process that involves complement pathway activity. We also quantified complement deposition on synapses in mouse visual thalamus and on cultured human neurons. Finally, we assessed uptake of synaptosomes by cultured microglia. We found that CSMD1 is present at synapses and interacts with complement proteins in the brain. Mice lacking Csmd1 displayed increased levels of complement component C3, an increased colocalization of C3 with presynaptic terminals, fewer retinogeniculate synapses, and aberrant segregation of eye-specific retinal inputs to the visual thalamus during the critical period of complement-dependent refinement of this circuit. Loss of CSMD1 in vivo enhanced synaptosome engulfment by microglia in vitro, and this effect was dependent on activity of the microglial complement receptor, CR3. Finally, human stem cell-derived neurons lacking CSMD1 were more vulnerable to complement deposition. These data suggest that CSMD1 can function as a regulator of complement-mediated synapse elimination in the brain during development.


Assuntos
Encéfalo , Proteínas de Membrana , Camundongos Knockout , Neurônios , Sinapses , Animais , Humanos , Camundongos , Encéfalo/metabolismo , Células Cultivadas , Complemento C3/metabolismo , Proteínas do Sistema Complemento/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Tálamo/metabolismo
4.
Nat Med ; 29(11): 2866-2884, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37814059

RESUMO

Huntington's disease (HD) is a devastating monogenic neurodegenerative disease characterized by early, selective pathology in the basal ganglia despite the ubiquitous expression of mutant huntingtin. The molecular mechanisms underlying this region-specific neuronal degeneration and how these relate to the development of early cognitive phenotypes are poorly understood. Here we show that there is selective loss of synaptic connections between the cortex and striatum in postmortem tissue from patients with HD that is associated with the increased activation and localization of complement proteins, innate immune molecules, to these synaptic elements. We also found that levels of these secreted innate immune molecules are elevated in the cerebrospinal fluid of premanifest HD patients and correlate with established measures of disease burden.In preclinical genetic models of HD, we show that complement proteins mediate the selective elimination of corticostriatal synapses at an early stage in disease pathogenesis, marking them for removal by microglia, the brain's resident macrophage population. This process requires mutant huntingtin to be expressed in both cortical and striatal neurons. Inhibition of this complement-dependent elimination mechanism through administration of a therapeutically relevant C1q function-blocking antibody or genetic ablation of a complement receptor on microglia prevented synapse loss, increased excitatory input to the striatum and rescued the early development of visual discrimination learning and cognitive flexibility deficits in these models. Together, our findings implicate microglia and the complement cascade in the selective, early degeneration of corticostriatal synapses and the development of cognitive deficits in presymptomatic HD; they also provide new preclinical data to support complement as a therapeutic target for early intervention.


Assuntos
Disfunção Cognitiva , Doença de Huntington , Doenças Neurodegenerativas , Humanos , Animais , Doença de Huntington/genética , Doenças Neurodegenerativas/patologia , Microglia/patologia , Sinapses/fisiologia , Corpo Estriado , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Proteína Huntingtina/genética , Proteínas do Sistema Complemento/metabolismo , Modelos Animais de Doenças
5.
Neuron ; 100(1): 120-134.e6, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30308165

RESUMO

Microglia regulate synaptic circuit remodeling and phagocytose synaptic material in the healthy brain; however, the mechanisms directing microglia to engulf specific synapses and avoid others remain unknown. Here, we demonstrate that an innate immune signaling pathway protects synapses from inappropriate removal. The expression patterns of CD47 and its receptor, SIRPα, correlated with peak pruning in the developing retinogeniculate system, and mice lacking these proteins exhibited increased microglial engulfment of retinogeniculate inputs and reduced synapse numbers in the dorsal lateral geniculate nucleus. CD47-deficient mice also displayed increased functional pruning, as measured by electrophysiology. In addition, CD47 was found to be required for neuronal activity-mediated changes in engulfment, as microglia in CD47 knockout mice failed to display preferential engulfment of less active inputs. Taken together, these results demonstrate that CD47-SIRPα signaling prevents excess microglial phagocytosis and show that molecular brakes can be regulated by activity to protect specific inputs.


Assuntos
Antígeno CD47/metabolismo , Microglia/metabolismo , Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/fisiologia , Receptores Imunológicos/metabolismo
6.
Nature ; 541(7638): 481-487, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28099414

RESUMO

Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood. Here we show that a subtype of reactive astrocytes, which we termed A1, is induced by classically activated neuroinflammatory microglia. We show that activated microglia induce A1 astrocytes by secreting Il-1α, TNF and C1q, and that these cytokines together are necessary and sufficient to induce A1 astrocytes. A1 astrocytes lose the ability to promote neuronal survival, outgrowth, synaptogenesis and phagocytosis, and induce the death of neurons and oligodendrocytes. Death of axotomized CNS neurons in vivo is prevented when the formation of A1 astrocytes is blocked. Finally, we show that A1 astrocytes are abundant in various human neurodegenerative diseases including Alzheimer's, Huntington's and Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. Taken together these findings help to explain why CNS neurons die after axotomy, strongly suggest that A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative disorders, and provide opportunities for the development of new treatments for these diseases.


Assuntos
Astrócitos/classificação , Astrócitos/patologia , Morte Celular , Sistema Nervoso Central/patologia , Microglia/patologia , Neurônios/patologia , Animais , Astrócitos/metabolismo , Axotomia , Técnicas de Cultura de Células , Sobrevivência Celular , Complemento C1q/metabolismo , Progressão da Doença , Humanos , Inflamação/patologia , Interleucina-1alfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neurodegenerativas/patologia , Oligodendroglia/patologia , Fagocitose , Fenótipo , Ratos , Ratos Sprague-Dawley , Sinapses/patologia , Toxinas Biológicas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Nature ; 534(7608): 538-43, 2016 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-27337340

RESUMO

Over 50% of patients who survive neuroinvasive infection with West Nile virus (WNV) exhibit chronic cognitive sequelae. Although thousands of cases of WNV-mediated memory dysfunction accrue annually, the mechanisms responsible for these impairments are unknown. The classical complement cascade, a key component of innate immune pathogen defence, mediates synaptic pruning by microglia during early postnatal development. Here we show that viral infection of adult hippocampal neurons induces complement-mediated elimination of presynaptic terminals in a murine WNV neuroinvasive disease model. Inoculation of WNV-NS5-E218A, a WNV with a mutant NS5(E218A) protein leads to survival rates and cognitive dysfunction that mirror human WNV neuroinvasive disease. WNV-NS5-E218A-recovered mice (recovery defined as survival after acute infection) display impaired spatial learning and persistence of phagocytic microglia without loss of hippocampal neurons or volume. Hippocampi from WNV-NS5-E218A-recovered mice with poor spatial learning show increased expression of genes that drive synaptic remodelling by microglia via complement. C1QA was upregulated and localized to microglia, infected neurons and presynaptic terminals during WNV neuroinvasive disease. Murine and human WNV neuroinvasive disease post-mortem samples exhibit loss of hippocampal CA3 presynaptic terminals, and murine studies revealed microglial engulfment of presynaptic terminals during acute infection and after recovery. Mice with fewer microglia (Il34(-/-) mice with a deficiency in IL-34 production) or deficiency in complement C3 or C3a receptor were protected from WNV-induced synaptic terminal loss. Our study provides a new murine model of WNV-induced spatial memory impairment, and identifies a potential mechanism underlying neurocognitive impairment in patients recovering from WNV neuroinvasive disease.


Assuntos
Proteínas do Sistema Complemento/imunologia , Transtornos da Memória/patologia , Transtornos da Memória/virologia , Microglia/imunologia , Plasticidade Neuronal , Terminações Pré-Sinápticas/patologia , Vírus do Nilo Ocidental/patogenicidade , Animais , Região CA3 Hipocampal/imunologia , Região CA3 Hipocampal/patologia , Região CA3 Hipocampal/virologia , Ativação do Complemento , Via Clássica do Complemento/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Transtornos da Memória/imunologia , Transtornos da Memória/fisiopatologia , Camundongos , Neurônios/imunologia , Neurônios/patologia , Neurônios/virologia , Terminações Pré-Sinápticas/imunologia , Memória Espacial , Febre do Nilo Ocidental/patologia , Febre do Nilo Ocidental/fisiopatologia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/imunologia
8.
Science ; 352(6286): 712-716, 2016 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-27033548

RESUMO

Synapse loss in Alzheimer's disease (AD) correlates with cognitive decline. Involvement of microglia and complement in AD has been attributed to neuroinflammation, prominent late in disease. Here we show in mouse models that complement and microglia mediate synaptic loss early in AD. C1q, the initiating protein of the classical complement cascade, is increased and associated with synapses before overt plaque deposition. Inhibition of C1q, C3, or the microglial complement receptor CR3 reduces the number of phagocytic microglia, as well as the extent of early synapse loss. C1q is necessary for the toxic effects of soluble ß-amyloid (Aß) oligomers on synapses and hippocampal long-term potentiation. Finally, microglia in adult brains engulf synaptic material in a CR3-dependent process when exposed to soluble Aß oligomers. Together, these findings suggest that the complement-dependent pathway and microglia that prune excess synapses in development are inappropriately activated and mediate synapse loss in AD.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Complemento C1q/imunologia , Microglia/imunologia , Fagocitose/imunologia , Sinapses/imunologia , Sinapses/patologia , Peptídeos beta-Amiloides/imunologia , Animais , Região CA1 Hipocampal/imunologia , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Transtornos Cognitivos/imunologia , Transtornos Cognitivos/patologia , Complemento C1q/genética , Via Clássica do Complemento/imunologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases/imunologia , Potenciação de Longa Duração , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Placa Amiloide/imunologia , Sinaptofisina/imunologia , Regulação para Cima
9.
Neuron ; 67(2): 239-52, 2010 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-20670832

RESUMO

The Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is critically required for the synaptic recruitment of AMPA-type glutamate receptors (AMPARs) during both development and plasticity. However, the underlying mechanism is unknown. Using single-particle tracking of AMPARs, we show that CaMKII activation and postsynaptic translocation induce the synaptic trapping of AMPARs diffusing in the membrane. AMPAR immobilization requires both phosphorylation of the auxiliary subunit Stargazin and its binding to PDZ domain scaffolds. It does not depend on the PDZ binding domain of GluA1 AMPAR subunit nor its phosphorylation at Ser831. Finally, CaMKII-dependent AMPAR immobilization regulates short-term plasticity. Thus, NMDA-dependent Ca(2+) influx in the post-synapse triggers a CaMKII- and Stargazin-dependent decrease in AMPAR diffusional exchange at synapses that controls synaptic function.


Assuntos
Canais de Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Neurônios/metabolismo , Receptores de AMPA/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Animais Recém-Nascidos , Benzotiadiazinas/farmacologia , Benzilaminas/farmacologia , Cálcio/metabolismo , Células Cultivadas , Difusão , Proteína 4 Homóloga a Disks-Large , Estimulação Elétrica/métodos , Embrião de Mamíferos , Ativação Enzimática/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Modelos Biológicos , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/genética , Estatísticas não Paramétricas , Sulfonamidas/farmacologia , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...