Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 613(7942): 120-129, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36517604

RESUMO

Myelin is required for the function of neuronal axons in the central nervous system, but the mechanisms that support myelin health are unclear. Although macrophages in the central nervous system have been implicated in myelin health1, it is unknown which macrophage populations are involved and which aspects they influence. Here we show that resident microglia are crucial for the maintenance of myelin health in adulthood in both mice and humans. We demonstrate that microglia are dispensable for developmental myelin ensheathment. However, they are required for subsequent regulation of myelin growth and associated cognitive function, and for preservation of myelin integrity by preventing its degeneration. We show that loss of myelin health due to the absence of microglia is associated with the appearance of a myelinating oligodendrocyte state with altered lipid metabolism. Moreover, this mechanism is regulated through disruption of the TGFß1-TGFßR1 axis. Our findings highlight microglia as promising therapeutic targets for conditions in which myelin growth and integrity are dysregulated, such as in ageing and neurodegenerative disease2,3.


Assuntos
Sistema Nervoso Central , Microglia , Bainha de Mielina , Adulto , Animais , Humanos , Camundongos , Axônios/metabolismo , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Microglia/citologia , Microglia/metabolismo , Microglia/patologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Cognição , Fator de Crescimento Transformador beta1/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Metabolismo dos Lipídeos , Envelhecimento/metabolismo , Envelhecimento/patologia
3.
Glia ; 72(2): 375-395, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37909242

RESUMO

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.


Assuntos
Transtornos Cerebrovasculares , Transtornos Cognitivos , Disfunção Cognitiva , Leucoencefalopatias , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos , Substância Branca , Animais , Camundongos , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/patologia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/patologia , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Leucoencefalopatias/genética , Leucoencefalopatias/metabolismo , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Receptores de Fator Estimulador de Colônias/metabolismo , Substância Branca/patologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo
4.
Hum Mol Genet ; 28(4): 628-638, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30351356

RESUMO

Mutations in the collagen genes COL4A1 and COL4A2 cause Mendelian eye, kidney and cerebrovascular disease including intracerebral haemorrhage (ICH), and common collagen IV variants are a risk factor for sporadic ICH. COL4A1 and COL4A2 mutations cause endoplasmic reticulum (ER) stress and basement membrane (BM) defects, and recent data suggest an association of ER stress with ICH due to a COL4A2 mutation. However, the potential of ER stress as a therapeutic target for the multi-systemic COL4A1 pathologies remains unclear. We performed a preventative oral treatment of Col4a1 mutant mice with the chemical chaperone phenyl butyric acid (PBA), which reduced adult ICH. Importantly, treatment of adult mice with the established disease also reduced ICH. However, PBA treatment did not alter eye and kidney defects, establishing tissue-specific outcomes of targeting Col4a1-derived ER stress, and therefore this treatment may not be applicable for patients with eye and renal disease. While PBA treatment reduced ER stress and increased collagen IV incorporation into BMs, the persistence of defects in BM structure and reduced ability of the BM to withstand mechanical stress indicate that PBA may be counter-indicative for pathologies caused by matrix defects. These data establish that treatment for COL4A1 disease requires a multipronged treatment approach that restores both ER homeostasis and matrix defects. Alleviating ER stress is a valid therapeutic target for preventing and treating established adult ICH, but collagen IV patients will require stratification based on their clinical presentation and mechanism of their mutations.


Assuntos
Hemorragia Cerebral/tratamento farmacológico , Colágeno Tipo IV/genética , Terapia de Alvo Molecular , Animais , Membrana Basal/efeitos dos fármacos , Membrana Basal/patologia , Hemorragia Cerebral/genética , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Camundongos , Mutação , Fenilbutiratos/administração & dosagem
5.
J Neuroinflammation ; 17(1): 367, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33261626

RESUMO

BACKGROUND: Chronic cerebral hypoperfusion causes damage to the brain's white matter underpinning vascular cognitive impairment. Inflammation and oxidative stress have been proposed as key pathophysiological mechanisms of which the transcription factor Nrf2 is a master regulator. We hypothesised that white matter pathology, microgliosis, blood-brain barrier breakdown and behavioural deficits induced by chronic hypoperfusion would be exacerbated in mice deficient in the transcription factor Nrf2. METHODS: Mice deficient in Nrf2 (male heterozygote or homozygous for Nrf2 knockout) or wild-type littermates on a C57Bl6/J background underwent bilateral carotid artery stenosis (BCAS) to induce chronic cerebral hypoperfusion or sham surgery and survived for a further 6 weeks. White matter pathology was assessed with MAG immunohistochemistry as a marker of altered axon-glial integrity; alterations to astrocytes and microglia/macrophages were assessed with GFAP and Iba1 immunohistochemistry, and blood-brain barrier breakdown was assessed with IgG immunohistochemistry. Behavioural alterations were assessed using 8-arm radial arm maze, and alterations to Nrf2-related and inflammatory-related genes were assessed with qRT-PCR. RESULTS: Chronic cerebral hypoperfusion induced white matter pathology, elevated microglial/macrophage levels and blood-brain barrier breakdown in white matter tracts that were increased in Nrf2+/- mice and further exacerbated by the complete absence of Nrf2. Chronic hypoperfusion induced white matter astrogliosis and induced an impairment in behaviour assessed with radial arm maze; however, these measures were not affected by Nrf2 deficiency. Although Nrf2-related antioxidant gene expression was not altered by chronic cerebral hypoperfusion, there was evidence for elevated pro-inflammatory related gene expression following chronic hypoperfusion that was not affected by Nrf2 deficiency. CONCLUSIONS: The results demonstrate that the absence of Nrf2 exacerbates white matter pathology and microgliosis following cerebral hypoperfusion but does not affect behavioural impairment.


Assuntos
Encéfalo/patologia , Disfunção Cognitiva/metabolismo , Microglia/patologia , Fator 2 Relacionado a NF-E2/deficiência , Substância Branca/patologia , Animais , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Estenose das Carótidas/complicações , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Glia ; 66(1): 34-46, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28722234

RESUMO

Chronic cerebral hypoperfusion is a key mechanism associated with white matter disruption in cerebral vascular disease and dementia. In a mouse model relevant to studying cerebral vascular disease, we have previously shown that cerebral hypoperfusion disrupts axon-glial integrity and the distribution of key paranodal and internodal proteins in subcortical myelinated axons. This disruption of myelinated axons is accompanied by increased microglia and cognitive decline. The aim of the present study was to investigate whether hypoperfusion impairs the functional integrity of white matter, its relation with axon-glial integrity and microglial number, and whether by targeting microglia these effects can be improved. We show that in response to increasing durations of hypoperfusion, the conduction velocity of myelinated fibres in the corpus callosum is progressively reduced and that paranodal and internodal axon-glial integrity is disrupted. The number of microglial cells increases in response to hypoperfusion and correlates with disrupted paranodal and internodal integrity and reduced conduction velocities. Further minocycline, a proposed anti-inflammatory and microglia inhibitor, restores white matter function related to a reduction in the number of microglia. The study suggests that microglial activation contributes to the structural and functional alterations of myelinated axons induced by cerebral hypoperfusion and that dampening microglia numbers/proliferation should be further investigated as potential therapeutic benefit in cerebral vascular disease.


Assuntos
Anti-Inflamatórios/uso terapêutico , Estenose das Carótidas , Gliose/tratamento farmacológico , Gliose/etiologia , Microglia/efeitos dos fármacos , Minociclina/uso terapêutico , Substância Branca/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Arginase/genética , Arginase/metabolismo , Axônios/patologia , Estenose das Carótidas/complicações , Estenose das Carótidas/tratamento farmacológico , Estenose das Carótidas/patologia , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/patologia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteína Associada a Mielina/metabolismo , Fibras Nervosas/efeitos dos fármacos , Fibras Nervosas/fisiologia , Substância Branca/patologia , Substância Branca/fisiologia
7.
J Neurochem ; 144(5): 617-633, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29210074

RESUMO

Advances in neuroimaging have enabled greater understanding of the progression of cerebral degenerative processes associated with ageing-related dementias. Leukoaraiosis or rarefied white matter (WM) originally described on computed tomography is one of the most prominent changes which occurs in older age. White matter hyperintensities (WMH) evident on magnetic resonance imaging have become commonplace to describe WM changes in relation to cognitive dysfunction, types of stroke injury, cerebral small vessel disease and neurodegenerative disorders including Alzheimer's disease. Substrates of WM degeneration collectively include myelin loss, axonal abnormalities, arteriolosclerosis and parenchymal changes resulting from lacunar infarcts, microinfarcts, microbleeds and perivascular spacing. WM cells incorporating astrocytes, oligodendrocytes, pericytes and microglia are recognized as key cellular components of the gliovascular unit. They respond to ongoing pathological processes in different ways leading to disruption of the gliovascular unit. The most robust alterations involve oligodendrocyte loss and astrocytic clasmatodendrosis with displacement of the water channel protein, aquaporin 4. These modifications likely precede arteriolosclerosis and capillary degeneration and involve tissue oedema, breach of the blood-brain barrier and induction of a chronic hypoxic state in the deep WM. Several pathophysiological mechanisms are proposed to explain how WM changes commencing with haemodynamic changes within the vascular system impact on cognitive dysfunction. Animal models simulating cerebral hypoperfusion in man have paved the way for several translational opportunities. Various compounds with variable efficacies have been tested to reduce oxidative stress, inflammation and blood-brain barrier damage in the WM. Our review demonstrates that WM degeneration encompasses multiple substrates and therefore more than one pharmacological approach is necessary to preserve axonal function and prevent cognitive impairment. This article is part of the Special Issue "Vascular Dementia".


Assuntos
Envelhecimento , Encéfalo/patologia , Demência Vascular/patologia , Demência/patologia , Leucoaraiose/complicações , Substância Branca/patologia , Animais , Barreira Hematoencefálica/patologia , Encéfalo/fisiopatologia , Demência/etiologia , Demência/fisiopatologia , Demência Vascular/etiologia , Demência Vascular/fisiopatologia , Humanos , Leucoaraiose/diagnóstico por imagem , Bainha de Mielina/patologia , Neuroglia/patologia , Neurônios/patologia , Substância Branca/fisiopatologia
8.
Clin Sci (Lond) ; 132(8): 851-868, 2018 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-29712883

RESUMO

Cerebral small vessel disease (SVD) is a major contributor to stroke, cognitive impairment and dementia with limited therapeutic interventions. There is a critical need to provide mechanistic insight and improve translation between pre-clinical research and the clinic. A 2-day workshop was held which brought together experts from several disciplines in cerebrovascular disease, dementia and cardiovascular biology, to highlight current advances in these fields, explore synergies and scope for development. These proceedings provide a summary of key talks at the workshop with a particular focus on animal models of cerebral vascular disease and dementia, mechanisms and approaches to improve translation. The outcomes of discussion groups on related themes to identify the gaps in knowledge and requirements to advance knowledge are summarized.


Assuntos
Doenças de Pequenos Vasos Cerebrais/etiologia , Pesquisa Translacional Biomédica , Animais , Humanos
9.
J Neuroinflammation ; 14(1): 81, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28399892

RESUMO

BACKGROUND: This study was designed to explore the beneficial effects of environmental enrichment (EE) on white matter glial changes in a mouse model of chronic cerebral hypoperfusion induced by bilateral common carotid artery stenosis (BCAS). METHODS: A total of 74 wild-type male C57BL/6J mice underwent BCAS or sham surgery. One week after surgery, the mice were randomly assigned into three different groups having varied amounts of EE-standard housing with no EE conditions (std), limited exposure with 3 h EE a day (3 h) and full-time exposure to EE (full) for 12 weeks. At 16 weeks after BCAS surgery, behavioural and cognitive function were assessed prior to euthanasia. Brain tissues were analysed for the degree of gliosis including morphological changes in astrocytes and microglia. RESULTS: Chronic cerebral hypoperfusion (or BCAS) increased clasmatodendrocytes (damaged astrocytes) with disruption of aquaporin-4 immunoreactivity and an increased degree of microglial activation/proliferation. BCAS also impaired behavioural and cognitive function. These changes were significantly attenuated, by limited exposure compared to full-time exposure to EE. CONCLUSIONS: Our results suggest that moderate or limited exposure to EE substantially reduced glial damage/activation. Our findings also suggest moderate rather than continuous exposure to EE is beneficial for patients with subcortical ischaemic vascular dementia characterised by white matter disease-related inflammation.


Assuntos
Estenose das Carótidas/complicações , Estenose das Carótidas/patologia , Meio Ambiente , Leucoencefalopatias/patologia , Neuroglia/metabolismo , Análise de Variância , Animais , Aquaporina 4/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/etiologia , Gliose/patologia , Leucoencefalopatias/etiologia , Leucoencefalopatias/enfermagem , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Comportamento de Nidação/fisiologia , Fatores de Tempo
10.
Clin Sci (Lond) ; 131(16): 2109-2123, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28667120

RESUMO

Cerebral hypoperfusion is an early feature of Alzheimer's disease (AD) that influences the progression from mild cognitive impairment to dementia. Understanding the mechanism is of critical importance in the search for new effective therapies. We hypothesized that cerebral hypoperfusion promotes the accumulation of amyloid-ß (Aß) and degenerative changes in the brain and is a potential mechanism contributing to development of dementia. To address this, we studied the effects of chronic cerebral hypoperfusion induced by bilateral carotid artery stenosis on Aß peptide pools in a transgenic mouse model of AD (transgenic mice with Swedish, Dutch and Iowa mutations in human amyloid precursor protein (APP) (Tg-SwDI)). Cerebrovascular integrity was characterized by quantifying the occurrence of microinfarcts and haemorrhages and compared with wild-type mice without Aß. A significant increase in soluble Aß peptides (Aß40/42) was detected after 1 month of hypoperfusion in the parenchyma in parallel with elevated APP and APP proteolytic products. Following 3 months, a significant increase in insoluble Aß40/42 was determined in the parenchyma and vasculature. Microinfarct load was significantly increased in the Tg-SwDI as compared with wild-type mice and further exacerbated by hypoperfusion at 1 and 3 months. In addition, the number of Tg-SwDI hypoperfused mice with haemorrhages was increased compared with hypoperfused wild-type mice. Soluble parenchymal Aß was associated with elevated NADPH oxidase-2 (NOX2) which was exacerbated by 1-month hypoperfusion. We suggest that in response to hypoperfusion, increased Aß production/deposition may contribute to degenerative processes by triggering oxidative stress promoting cerebrovascular disruption and the development of microinfarcts.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Isquemia Encefálica/complicações , Angiopatia Amiloide Cerebral/etiologia , Hemorragia Cerebral/etiologia , Infarto Cerebral/etiologia , Fragmentos de Peptídeos/metabolismo , Animais , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Angiopatia Amiloide Cerebral/metabolismo , Angiopatia Amiloide Cerebral/patologia , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patologia , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Circulação Cerebrovascular/fisiologia , Doença Crônica , Modelos Animais de Doenças , Glicoproteínas de Membrana/metabolismo , Camundongos Transgênicos , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Estresse Oxidativo/fisiologia , Tecido Parenquimatoso/metabolismo , Solubilidade
11.
Clin Sci (Lond) ; 131(19): 2451-2468, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28963120

RESUMO

Increasing evidence suggests that vascular risk factors contribute to neurodegeneration, cognitive impairment and dementia. While there is considerable overlap between features of vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD), it appears that cerebral hypoperfusion is the common underlying pathophysiological mechanism which is a major contributor to cognitive decline and degenerative processes leading to dementia. Sustained cerebral hypoperfusion is suggested to be the cause of white matter attenuation, a key feature common to both AD and dementia associated with cerebral small vessel disease (SVD). White matter changes increase the risk for stroke, dementia and disability. A major gap has been the lack of mechanistic insights into the evolution and progress of VCID. However, this gap is closing with the recent refinement of rodent models which replicate chronic cerebral hypoperfusion. In this review, we discuss the relevance and advantages of these models in elucidating the pathogenesis of VCID and explore the interplay between hypoperfusion and the deposition of amyloid ß (Aß) protein, as it relates to AD. We use examples of our recent investigations to illustrate the utility of the model in preclinical testing of candidate drugs and lifestyle factors. We propose that the use of such models is necessary for tackling the urgently needed translational gap from preclinical models to clinical treatments.


Assuntos
Comportamento Animal , Circulação Cerebrovascular , Transtornos Cerebrovasculares/complicações , Transtornos Cognitivos/etiologia , Cognição , Demência Vascular/etiologia , Pesquisa Translacional Biomédica/métodos , Peptídeos beta-Amiloides , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Transtornos Cerebrovasculares/tratamento farmacológico , Transtornos Cerebrovasculares/fisiopatologia , Transtornos Cerebrovasculares/psicologia , Doença Crônica , Cognição/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/fisiopatologia , Transtornos Cognitivos/psicologia , Demência Vascular/tratamento farmacológico , Demência Vascular/fisiopatologia , Demência Vascular/psicologia , Modelos Animais de Doenças , Progressão da Doença , Humanos , Leucoencefalopatias/etiologia , Leucoencefalopatias/fisiopatologia , Leucoencefalopatias/psicologia , Placa Amiloide , Fatores de Risco , Especificidade da Espécie , Fatores de Tempo
12.
Brain ; 139(Pt 1): 242-58, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26667280

RESUMO

White matter hyperintensities as seen on brain T2-weighted magnetic resonance imaging are associated with varying degrees of cognitive dysfunction in stroke, cerebral small vessel disease and dementia. The pathophysiological mechanisms within the white matter accounting for cognitive dysfunction remain unclear. With the hypothesis that gliovascular interactions are impaired in subjects with high burdens of white matter hyperintensities, we performed clinicopathological studies in post-stroke survivors, who had exhibited greater frontal white matter hyperintensities volumes that predicted shorter time to dementia onset. Histopathological methods were used to identify substrates in the white matter that would distinguish post-stroke demented from post-stroke non-demented subjects. We focused on the reactive cell marker glial fibrillary acidic protein (GFAP) to study the incidence and location of clasmatodendrosis, a morphological attribute of irreversibly injured astrocytes. In contrast to normal appearing GFAP+ astrocytes, clasmatodendrocytes were swollen and had vacuolated cell bodies. Other markers such as aldehyde dehydrogenase 1 family, member L1 (ALDH1L1) showed cytoplasmic disintegration of the astrocytes. Total GFAP+ cells in both the frontal and temporal white matter were not greater in post-stroke demented versus post-stroke non-demented subjects. However, the percentage of clasmatodendrocytes was increased by >2-fold in subjects with post-stroke demented compared to post-stroke non-demented subjects (P = 0.026) and by 11-fold in older controls versus young controls (P < 0.023) in the frontal white matter. High ratios of clasmotodendrocytes to total astrocytes in the frontal white matter were consistent with lower Mini-Mental State Examination and the revised Cambridge Cognition Examination scores in post-stroke demented subjects. Double immunofluorescent staining showed aberrant co-localization of aquaporin 4 (AQP4) in retracted GFAP+ astrocytes with disrupted end-feet juxtaposed to microvessels. To explore whether this was associated with the disrupted gliovascular interactions or blood-brain barrier damage, we assessed the co-localization of GFAP and AQP4 immunoreactivities in post-mortem brains from adult baboons with cerebral hypoperfusive injury, induced by occlusion of three major vessels supplying blood to the brain. Analysis of the frontal white matter in perfused brains from the animals surviving 1-28 days after occlusion revealed that the highest intensity of fibrinogen immunoreactivity was at 14 days. At this survival time point, we also noted strikingly similar redistribution of AQP4 and GFAP+ astrocytes transformed into clasmatodendrocytes. Our findings suggest novel associations between irreversible astrocyte injury and disruption of gliovascular interactions at the blood-brain barrier in the frontal white matter and cognitive impairment in elderly post-stroke survivors. We propose that clasmatodendrosis is another pathological substrate, linked to white matter hyperintensities and frontal white matter changes, which may contribute to post-stroke or small vessel disease dementia.


Assuntos
Envelhecimento/patologia , Astrócitos/patologia , Demência/complicações , Demência/patologia , Acidente Vascular Cerebral/complicações , Substância Branca/patologia , Idoso , Idoso de 80 Anos ou mais , Aldeído Desidrogenase/metabolismo , Animais , Aquaporina 4/metabolismo , Astrócitos/metabolismo , Barreira Hematoencefálica/patologia , Estudos de Casos e Controles , Transtornos Cognitivos/patologia , Feminino , Lobo Frontal/irrigação sanguínea , Lobo Frontal/patologia , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Neuroimagem , Testes Neuropsicológicos , Oxirredutases atuantes sobre Doadores de Grupo CH-NH , Papio anubis , Acidente Vascular Cerebral/patologia , Substância Branca/irrigação sanguínea
13.
Acta Neuropathol Commun ; 12(1): 29, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38360798

RESUMO

Neurovascular unit mural cells called 'pericytes' maintain the blood-brain barrier and local cerebral blood flow. Pathological changes in the hippocampus predispose to cognitive impairment and dementia. The role of hippocampal pericytes in dementia is largely unknown. We investigated hippocampal pericytes in 90 post-mortem brains from post-stroke dementia (PSD), vascular dementia (VaD), Alzheimer's disease (AD), and AD-VaD (Mixed) subjects, and post-stroke non-demented survivors as well as similar age controls. We used collagen IV immunohistochemistry to determine pericyte densities and a mouse model of VaD to validate the effects of chronic cerebral hypoperfusion. Despite increased trends in hippocampal microvascular densities across all dementias, mean pericyte densities were reduced by ~25-40% in PSD, VaD and AD subjects compared to those in controls, which calculated to 14.1 ± 0.7 per mm capillary length, specifically in the cornu ammonis (CA) 1 region (P = 0.01). In mice with chronic bilateral carotid artery occlusion, hippocampal pericyte loss was ~60% relative to controls (P < 0.001). Pericyte densities were correlated with CA1 volumes (r = 0.54, P = 0.006) but not in any other sub-region. However, mice subjected to the full-time environmental enrichment (EE) paradigm showed remarkable attenuation of hippocampal CA1 pericyte loss in tandem with CA1 atrophy. Our results suggest loss of hippocampal microvascular pericytes across common dementias is explained by a vascular aetiology, whilst the EE paradigm offers significant protection.


Assuntos
Doença de Alzheimer , Isquemia Encefálica , Demência Vascular , Acidente Vascular Cerebral , Humanos , Camundongos , Animais , Doença de Alzheimer/patologia , Demência Vascular/patologia , Pericitos/patologia , Hipocampo/patologia , Encéfalo/patologia , Acidente Vascular Cerebral/patologia , Isquemia Encefálica/patologia
14.
Cell Rep ; 43(5): 114250, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38762882

RESUMO

Acute stroke triggers extensive changes to myeloid immune cell populations in the brain that may be targets for limiting brain damage and enhancing repair. Immunomodulatory approaches will be most effective with precise manipulation of discrete myeloid cell phenotypes in time and space. Here, we investigate how stroke alters mononuclear myeloid cell composition and phenotypes at single-cell resolution and key spatial patterns. Our results show that multiple reactive microglial states and monocyte-derived populations contribute to an extensive myeloid cell repertoire in post-stroke brains. We identify important overlaps and distinctions among different cell types/states that involve ontogeny- and spatial-related properties. Notably, brain connectivity with infarcted tissue underpins the pattern of local and remote altered cell accumulation and reactivity. Our discoveries suggest a global but anatomically governed brain myeloid cell response to stroke that comprises diverse phenotypes arising through intrinsic cell ontogeny factors interacting with exposure to spatially organized brain damage and neuro-axonal cues.


Assuntos
Encéfalo , Microglia , Células Mieloides , Fenótipo , Acidente Vascular Cerebral , Animais , Encéfalo/patologia , Acidente Vascular Cerebral/patologia , Células Mieloides/metabolismo , Microglia/patologia , Microglia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Monócitos/patologia
15.
Hum Mol Genet ; 20(12): 2406-21, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21478199

RESUMO

Apolipoprotein E (apoE) is a 34 kDa glycoprotein with three distinct isoforms in the human population (apoE2, apoE3 and apoE4) known to play a major role in differentially influencing risk to, as well as outcome from, disease and injury in the central nervous system. In general, the apoE4 allele is associated with poorer outcomes after disease or injury, whereas apoE3 is associated with better responses. The extent to which different apoE isoforms influence degenerative and regenerative events in the peripheral nervous system (PNS) is still to be established, and the mechanisms through which apoE exerts its isoform-specific effects remain unclear. Here, we have investigated isoform-specific effects of human apoE on the mouse PNS. Experiments in mice ubiquitously expressing human apoE3 or human apoE4 on a null mouse apoE background revealed that apoE4 expression significantly disrupted peripheral nerve regeneration and subsequent neuromuscular junction re-innervation following nerve injury compared with apoE3, with no observable effects on normal development, maturation or Wallerian degeneration. Proteomic isobaric tag for relative and absolute quantitation (iTRAQ) screens comparing healthy and regenerating peripheral nerves from mice expressing apoE3 or apoE4 revealed significant differences in networks of proteins regulating cellular outgrowth and regeneration (myosin/actin proteins), as well as differences in expression levels of proteins involved in regulating the blood-nerve barrier (including orosomucoid 1). Taken together, these findings have identified isoform-specific roles for apoE in determining the protein composition of peripheral nerve as well as regulating nerve regeneration pathways in vivo.


Assuntos
Apolipoproteínas E/metabolismo , Regeneração Nervosa/fisiologia , Sistema Nervoso Periférico/fisiologia , Isoformas de Proteínas/metabolismo , Animais , Apolipoproteínas E/genética , Axônios/metabolismo , Axônios/ultraestrutura , Western Blotting , Eletrofisiologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Orosomucoide/metabolismo , Sistema Nervoso Periférico/lesões , Isoformas de Proteínas/genética , Proteômica/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem
16.
Stem Cell Reports ; 18(12): 2386-2399, 2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-37977146

RESUMO

Cerebral small vessel disease (SVD) affects the small vessels in the brain and is a leading cause of stroke and dementia. Emerging evidence supports a role of the extracellular matrix (ECM), at the interface between blood and brain, in the progression of SVD pathology, but this remains poorly characterized. To address ECM role in SVD, we developed a co-culture model of mural and endothelial cells using human induced pluripotent stem cells from patients with COL4A1/A2 SVD-related mutations. This model revealed that these mutations induce apoptosis, migration defects, ECM remodeling, and transcriptome changes in mural cells. Importantly, these mural cell defects exert a detrimental effect on endothelial cell tight junctions through paracrine actions. COL4A1/A2 models also express high levels of matrix metalloproteinases (MMPs), and inhibiting MMP activity partially rescues the ECM abnormalities and mural cell phenotypic changes. These data provide a basis for targeting MMP as a therapeutic opportunity in SVD.


Assuntos
Células-Tronco Pluripotentes Induzidas , Acidente Vascular Cerebral , Humanos , Células Endoteliais , Encéfalo/patologia , Acidente Vascular Cerebral/patologia , Matriz Extracelular , Metaloproteinases da Matriz/genética , Colágeno Tipo IV/genética
17.
J Neurosci ; 31(49): 18185-94, 2011 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22159130

RESUMO

Myelinated axons have a distinct protein architecture essential for action potential propagation, neuronal communication, and maintaining cognitive function. Damage to myelinated axons, associated with cerebral hypoperfusion, contributes to age-related cognitive decline. We sought to determine early alterations in the protein architecture of myelinated axons and potential mechanisms after hypoperfusion. Using a mouse model of hypoperfusion, we assessed changes in proteins critical to the maintenance of paranodes, nodes of Ranvier, axon-glial integrity, axons, and myelin by confocal laser scanning microscopy. As early as 3 d after hypoperfusion, the paranodal septate-like junctions were damaged. This was marked by a progressive reduction of paranodal Neurofascin signal and a loss of septate-like junctions. Concurrent with paranodal disruption, there was a significant increase in nodal length, identified by Nav1.6 staining, with hypoperfusion. Disruption of axon-glial integrity was also determined after hypoperfusion by changes in the spatial distribution of myelin-associated glycoprotein staining. These nodal/paranodal changes were more pronounced after 1 month of hypoperfusion. In contrast, the nodal anchoring proteins AnkyrinG and Neurofascin 186 were unchanged and there were no overt changes in axonal and myelin integrity with hypoperfusion. A microarray analysis of white matter samples indicated that there were significant alterations in 129 genes. Subsequent analysis indicated alterations in biological pathways, including inflammatory responses, cytokine-cytokine receptor interactions, blood vessel development, and cell proliferation processes. Our results demonstrate that hypoperfusion leads to a rapid disruption of key proteins critical to the stability of the axon-glial connection that is mediated by a diversity of molecular events.


Assuntos
Axônios/patologia , Regulação da Expressão Gênica/fisiologia , Hipóxia-Isquemia Encefálica/patologia , Neuroglia/patologia , Neurônios/patologia , Fatores Etários , Animais , Anquirinas/metabolismo , Moléculas de Adesão Celular , Moléculas de Adesão Celular Neuronais , Doença Crônica , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Modelos Animais de Doenças , Tomografia com Microscopia Eletrônica/métodos , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Proteína Básica da Mielina/metabolismo , Glicoproteína Associada a Mielina/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.6 , Fibras Nervosas Mielinizadas/metabolismo , Fatores de Crescimento Neural , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neuroglia/metabolismo , Neurônios/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Nervo Óptico/ultraestrutura , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/patologia , Transdução de Sinais/fisiologia , Canais de Sódio
18.
Nat Neurosci ; 11(4): 476-87, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18344994

RESUMO

Intrinsic antioxidant defenses are important for neuronal longevity. We found that in rat neurons, synaptic activity, acting via NMDA receptor (NMDAR) signaling, boosted antioxidant defenses by making changes to the thioredoxin-peroxiredoxin (Prx) system. Synaptic activity enhanced thioredoxin activity, facilitated the reduction of overoxidized Prxs and promoted resistance to oxidative stress. Resistance was mediated by coordinated transcriptional changes; synaptic NMDAR activity inactivated a previously unknown Forkhead box O target gene, the thioredoxin inhibitor Txnip. Conversely, NMDAR blockade upregulated Txnip in vivo and in vitro, where it bound thioredoxin and promoted vulnerability to oxidative damage. Synaptic activity also upregulated the Prx reactivating genes Sesn2 (sestrin 2) and Srxn1 (sulfiredoxin), via C/EBPbeta and AP-1, respectively. Mimicking these expression changes was sufficient to strengthen antioxidant defenses. Trans-synaptic stimulation of synaptic NMDARs was crucial for boosting antioxidant defenses; chronic bath activation of all (synaptic and extrasynaptic) NMDARs induced no antioxidative effects. Thus, synaptic NMDAR activity may influence the progression of pathological processes associated with oxidative damage.


Assuntos
Antioxidantes/metabolismo , Estresse Oxidativo/fisiologia , Peroxirredoxinas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Tiorredoxinas/metabolismo , Animais , Proteínas de Transporte/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica/fisiologia , Camundongos , Neurônios/metabolismo , Proteínas Nucleares , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Peroxidases , Proteínas/metabolismo , Ratos , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Transcrição Gênica/fisiologia
19.
J Cereb Blood Flow Metab ; 42(7): 1176-1191, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35102790

RESUMO

Chronic microvascular inflammation and oxidative stress are inter-related mechanisms underpinning white matter disease and vascular cognitive impairment (VCI). A proposed mediator is nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2), a major source of reactive oxygen species (ROS) in the brain. To assess the role of Nox2 in VCI, we studied a tractable model with white matter pathology and cognitive impairment induced by bilateral carotid artery stenosis (BCAS). Mice with genetic deletion of Nox2 (Nox2 KO) were compared to wild-type (WT) following BCAS. Sustained BCAS over 12 weeks in WT mice induced Nox2 expression, indices of microvascular inflammation and oxidative damage, along with white matter pathology culminating in a marked cognitive impairment, which were all protected by Nox2 genetic deletion. Neurovascular coupling was impaired in WT mice post-BCAS and restored in Nox2 KO mice. Increased vascular expression of chemoattractant mediators, cell-adhesion molecules and endothelial activation factors in WT mice post-BCAS were ameliorated by Nox2 deficiency. The clinical relevance was confirmed by increased vascular Nox2 and indices of microvascular inflammation in human post-mortem subjects with cerebral vascular disease. Our results support Nox2 activity as a critical determinant of VCI, whose targeting may be of therapeutic benefit in cerebral vascular disease.


Assuntos
Estenose das Carótidas , Disfunção Cognitiva , NADPH Oxidase 2 , Substância Branca , Animais , Disfunção Cognitiva/patologia , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo , Substância Branca/patologia
20.
Brain Pathol ; 32(6): e13101, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35748290

RESUMO

With the hypothesis that perivascular microglia are involved as neuroinflammatory components of the gliovascular unit contributing to white matter hyperintensities on MRI and pathophysiology, we assessed their status in stroke survivors who develop dementia. Immunohistochemical and immunofluorescent methods were used to assess the distribution and quantification of total and perivascular microglial cell densities in 68 brains focusing on the frontal lobe WM and overlying neocortex in post-stroke dementia (PSD), post-stroke non-dementia (PSND) and similar age control subjects. We primarily used CD68 as a marker of phagocytic microglia, as well as other markers of microglia including Iba-1 and TMEM119, and the myeloid cell marker TREM2 to assess dementia-specific changes. We first noted greater total densities of CD68+ and TREM2+ cells per mm2 in the frontal WM compared to the overlying cortex across the stroke cases and controls (p = 0.001). PSD subjects showed increased percentage of activated perivascular CD68+ cells distinct from ramified or primed microglia in the WM (p < 0.05). However, there was no apparent change in perivascular TREM2+ cells. Total densities of TREM2+ cells were only ~10% of CD68+ cells but there was high degree of overlap (>70%) between them in both the WM and the cortex. CD68 and Iba-1 or CD68 and TMEM119 markers were colocalised by ~55%. Within the deep WM, ~30% of CD68+ cells were co-localised with fragments of degraded myelin basic protein. Among fragmented CD68+ cells in adjacent WM of PSD subjects, >80% of the cells expressed cleaved caspase-3. Our observations suggest although the overall repertoire of perivascular microglial cells is not changed in the parenchyma, PSD subjects accrue more perivascular-activated CD68+ microglia rather than TREM2+ cells. This implies there is a subset of CD68+ cells, which are responsible for the differential response in perivascular inflammation within the gliovascular unit of the deep WM.


Assuntos
Demência Vascular , Acidente Vascular Cerebral , Substância Branca , Humanos , Demência Vascular/metabolismo , Microglia/metabolismo , Encéfalo , Acidente Vascular Cerebral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA