Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Obes Metab Syndr ; 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38736362

RESUMO

Intermittent fasting (IF), a dietary pattern alternating between eating and fasting periods within a 24-hour cycle, has garnered recognition for its potential to enhance both healthspan and lifespan in animal models and humans. It also shows promise in alleviating age-related diseases, including neurodegeneration. Vascular cognitive impairment (VCI) spans a severity range from mild cognitive deficits to severe cognitive deficits and loss of function in vascular dementia. Chronic cerebral hypoperfusion has emerged as a significant contributor to VCI, instigating vascular pathologies such as microbleeds, blood-brain barrier dysfunction, neuronal loss and white matter lesions. Preclinical studies in rodents strongly suggest that IF has the potential to attenuate pathological mechanisms, including excitotoxicity, oxidative stress, inflammation, and cell death pathways in VCI models. Hence, this supports evaluating IF in clinical trials for both existing and at-risk VCI patients. This review compiles existing data supporting IF's potential in treating VCI-related vascular and neuronal pathologies, emphasizing the mechanisms by which IF may mitigate these issues. Hence providing a comprehensive overview of the available data supporting IF's potential in treating VCI by emphasizing the underlying mechanisms that make IF a promising intervention for VCI.

2.
Elife ; 122023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37769126

RESUMO

Intermittent fasting (IF) has been shown to reduce cardiovascular risk factors in both animals and humans, and can protect the heart against ischemic injury in models of myocardial infarction. However, the underlying molecular mechanisms behind these effects remain unclear. To shed light on the molecular and cellular adaptations of the heart to IF, we conducted comprehensive system-wide analyses of the proteome, phosphoproteome, and transcriptome, followed by functional analysis. Using advanced mass spectrometry, we profiled the proteome and phosphoproteome of heart tissues obtained from mice that were maintained on daily 12- or 16 hr fasting, every-other-day fasting, or ad libitum control feeding regimens for 6 months. We also performed RNA sequencing to evaluate whether the observed molecular responses to IF occur at the transcriptional or post-transcriptional levels. Our analyses revealed that IF significantly affected pathways that regulate cyclic GMP signaling, lipid and amino acid metabolism, cell adhesion, cell death, and inflammation. Furthermore, we found that the impact of IF on different metabolic processes varied depending on the length of the fasting regimen. Short IF regimens showed a higher correlation of pathway alteration, while longer IF regimens had an inverse correlation of metabolic processes such as fatty acid oxidation and immune processes. Additionally, functional echocardiographic analyses demonstrated that IF enhances stress-induced cardiac performance. Our systematic multi-omics study provides a molecular framework for understanding how IF impacts the heart's function and its vulnerability to injury and disease.


Assuntos
Jejum Intermitente , Multiômica , Humanos , Camundongos , Animais , Proteoma , Jejum/fisiologia , Metabolismo Energético
4.
Neuromolecular Med ; 25(4): 533-544, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37646911

RESUMO

Inflammation is a hallmark mechanism of ischemic stroke-induced brain injury. Recent studies have shown that an intracellular multimeric protein complex known as an inflammasome is a key factor for inducing an inflammatory response, and apoptotic and pyroptotic cell death in ischemic stroke. Inflammasome assembly leads to the activation of pro-inflammatory caspases, and the maturation and secretion of pro-inflammatory cytokines IL-1ß and IL-18. While the role of inflammasomes in ischemic stroke-induced neuronal death, and microglial activation and cell death have been established, little is known about the role of inflammasomes in astrocytes under ischemic conditions. In this study, we investigated the expression and activation of inflammasome components in protoplasmic and fibrous astrocytes under ischemic conditions. We found that both protoplasmic and fibrous astrocytes expressed a differential increase in inflammasome protein components, and that their activation promoted maturation of IL-1ß and IL-18, and secretion of IL-1ß, as well as initiating apoptotic and pyroptotic cell death. Pharmacological inhibition of caspase-1 decreased expression of cleaved caspase-1 and production of mature IL-1ß, and protected against inflammasome-mediated apoptotic and pyroptotic cell death. Overall, this study provides novel insights into the role of inflammasome signaling in astrocytes under ischemic conditions.


Assuntos
Inflamassomos , AVC Isquêmico , Humanos , Interleucina-18 , Astrócitos/metabolismo , Caspase 1/metabolismo , Caspases/metabolismo , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
5.
Acta Neuropathol Commun ; 11(1): 93, 2023 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-37309012

RESUMO

Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.


Assuntos
Isquemia Encefálica , Transtornos Cognitivos , Disfunção Cognitiva , Humanos , Circulação Cerebrovascular , Neuropatologia
6.
J Cereb Blood Flow Metab ; 43(5): 722-735, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36537035

RESUMO

Chronic cerebral hypoperfusion (CCH) is postulated to underlie multiple pathophysiological processes in vascular dementia (VaD), including extracellular matrix dysfunction. While several extracellular matrix proteins, namely cyclophilin A (CypA), extracellular matrix metalloproteinase inducer (EMMPRIN) and gelatinases (matrix metalloproteinases, MMP-2 and -9) have been investigated in acute stroke, their involvement in CCH and VaD remains unclear. In this study, CypA-EMMPRIN-gelatinase proteins were analysed in a clinical cohort of 36 aged, cognitively unimpaired subjects and 48 VaD patients, as well as in a bilateral carotid artery stenosis mouse model of CCH. Lower CypA and higher EMMPRIN levels were found in both VaD serum and CCH mouse brain. Furthermore, gelatinases were differentially altered in CCH mice and VaD patients, with significant MMP-2 increase in CCH brain and serum, whilst serum MMP-9 was elevated in VaD but reduced in CCH, suggesting complex CypA-EMMPRIN-gelatinase regulatory mechanisms. Interestingly, subjects with cortical infarcts had higher serum MMP-2, while white matter hyperintensities, cortical infarcts and lacunes were associated with higher serum MMP-9. Taken together, our data indicate that perturbations of CypA-EMMPRIN signalling may be associated with gelatinase-mediated vascular sequelae, highlighting the potential utility of the CypA-EMMPRIN-gelatinase pathway as clinical biomarkers and therapeutic targets in VaD.


Assuntos
Isquemia Encefálica , Demência Vascular , Camundongos , Animais , Basigina/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinase 2 da Matriz , Ciclofilina A/metabolismo , Gelatinases , Infarto
7.
Int J Biol Sci ; 18(16): 6052-6067, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36439869

RESUMO

Background - Chronic cerebral hypoperfusion (CCH) is an important pathophysiological mechanism of vascular cognitive impairment (VCI). The heterogeneous effects of CCH complicate establishing single target therapies against VCI and its more severe form, vascular dementia (VaD). Intermittent fasting (IF) has multiple targets and is neuroprotective across a range of disease conditions including stroke, but its effects against CCH-induced neurovascular pathologies remain to be elucidated. We therefore assessed the effect of IF against CCH-associated neurovascular pathologies and investigated its underlying mechanisms. Methods - Male C57BL/6NTac mice were subjected to either ad libitum feeding (AL) or IF (16 hours of fasting per day) for 4 months. In both groups, CCH was experimentally induced by the bilateral common carotid artery stenosis (BCAS) method. Sham operated groups were used as controls. Measures of leaky microvessels, blood-brain barrier (BBB) permeability, protein expression of tight junctions, extracellular matrix components and white matter changes were determined to investigate the effect of IF against CCH-induced neurovascular pathologies. Results - IF alleviated CCH-induced neurovascular pathologies by reducing the number of leaky microvessels, BBB breakdown and loss of tight junctional proteins. In addition, IF mitigated the severity of white matter lesions, and maintained myelin basic protein levels, while concurrently reducing hippocampal neuronal cell death. Furthermore, IF reduced the CCH-induced increase in levels of matrix metalloproteinase (MMP)-2 and its upstream activator MT1-MMP, which are involved in the breakdown of the extracellular matrix that is a core component of the BBB. Additionally, we observed that IF reduced CCH-induced increase in the oxidative stress marker malondialdehyde, and increased antioxidant markers glutathione and superoxide dismutase. Overall, our data suggest that IF attenuates neurovascular damage, metalloproteinase and oxidative stress-associated pathways, and cell death in the brain following CCH in a mouse model of VCI. Conclusion - Although IF has yet to be assessed in human patients with VaD, our data suggest that IF may be an effective means of preventing the onset or suppressing the development of neurovascular pathologies in VCI and VaD.


Assuntos
Isquemia Encefálica , Estenose das Carótidas , Disfunção Cognitiva , Animais , Camundongos , Humanos , Masculino , Jejum , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Neurônios/metabolismo , Isquemia Encefálica/metabolismo , Estenose das Carótidas/complicações , Estenose das Carótidas/patologia
8.
Theranostics ; 12(4): 1639-1658, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35198062

RESUMO

The prevalence of cerebrovascular disease increases with age, placing the elderly at a greater lifetime risk for dementia. Vascular cognitive impairment (VCI) encompasses a spectrum of cognitive deficits from mild cognitive impairment to dementia. VCI and its most severe form, vascular dementia (VaD), is becoming a major public health concern worldwide. As growing efforts are being taken to understand VCI and VaD in animal models and humans, the pathogenesis of the disease is being actively explored. It is postulated that chronic cerebral hypoperfusion (CCH) is a major cause of VCI. CCH activates a molecular and cellular injury cascade that leads to breakdown of the blood brain barrier (BBB) and neurodegeneration. The BBB tightly regulates the movement of substances between the blood and the brain, thereby regulating the microenvironment within the brain parenchyma. Here we illustrate how BBB damage is causal in the pathogenesis of VCI through the increased activation of pathways related to excitotoxicity, oxidative stress, inflammation and matrix metalloproteinases that lead to downstream perivascular damage, leukocyte infiltration and white matter changes in the brain. Thus, CCH-induced BBB damage may initiate and contribute to a vicious cycle, resulting in progressive neuropathological changes of VCI in the brain. This review outlines the molecular and cellular mechanisms that govern BBB breakdown during CCH and highlights the clinical evidence in identifying at-risk VCI patients.


Assuntos
Isquemia Encefálica , Disfunção Cognitiva , Demência Vascular , Idoso , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Isquemia Encefálica/patologia , Disfunção Cognitiva/metabolismo , Demência Vascular/etiologia , Demência Vascular/metabolismo , Humanos
9.
Neurochem Int ; 148: 105109, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34174333

RESUMO

Chronic cerebral hypoperfusion (CCH) has been shown to initiate several inflammatory pathways that can contribute to cognitive deficits and memory loss in vascular cognitive impairment (VCI). Multi-protein complexes termed inflammasomes that may be involved in the inflammatory response to CCH has already been shown to contribute to the inflammatory process and cell death following acute cerebral ischemia. Intermittent fasting (IF) has already been shown to decrease inflammasome activation and protect the brain from ischemic stroke; however, its effects during CCH remains unknown. The present study investigated the impact of IF (16 h of food deprivation daily) for four months on inflammasome-mediated cell death in the cerebellum following CCH in a mouse model of VCI using fourteen to sixteen-week-old male C57BL/6NTac mice. Here we demonstrated that IF decreased inflammasome activation, and initiation of apoptotic and pyroptotic cell death pathways as reflected by the reduction (20-30%) in the expression levels of key effector proteins and cell death markers in the cerebellum following CCH. In summary, our results indicate that IF can attenuate the inflammatory response and cell death pathways in the brain following chronic hypoperfusion in a mouse model of VCI.


Assuntos
Apoptose/fisiologia , Isquemia Encefálica/patologia , Isquemia Encefálica/prevenção & controle , Encéfalo/patologia , Jejum , Inflamassomos , Piroptose/fisiologia , Animais , Isquemia Encefálica/psicologia , Estenose das Carótidas/patologia , Cerebelo/patologia , Disfunção Cognitiva , Demência Vascular/patologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL
10.
Mol Psychiatry ; 26(8): 4544-4560, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33299135

RESUMO

Chronic cerebral hypoperfusion is associated with vascular dementia (VaD). Cerebral hypoperfusion may initiate complex molecular and cellular inflammatory pathways that contribute to long-term cognitive impairment and memory loss. Here we used a bilateral common carotid artery stenosis (BCAS) mouse model of VaD to investigate its effect on the innate immune response-particularly the inflammasome signaling pathway. Comprehensive analyses revealed that chronic cerebral hypoperfusion induces a complex temporal expression and activation of inflammasome components and their downstream products (IL-1ß and IL-18) in different brain regions, and promotes activation of apoptotic and pyroptotic cell death pathways. Polarized glial-cell activation, white-matter lesion formation and hippocampal neuronal loss also occurred in a spatiotemporal manner. Moreover, in AIM2 knockout mice we observed attenuated inflammasome-mediated production of proinflammatory cytokines, apoptosis, and pyroptosis, as well as resistance to chronic microglial activation, myelin breakdown, hippocampal neuronal loss, and behavioral and cognitive deficits following BCAS. Hence, we have demonstrated that activation of the AIM2 inflammasome substantially contributes to the pathophysiology of chronic cerebral hypoperfusion-induced brain injury and may therefore represent a promising therapeutic target for attenuating cognitive impairment in VaD.


Assuntos
Disfunção Cognitiva , Demência Vascular , Substância Branca , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Inflamassomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Substância Branca/metabolismo
11.
Neuromolecular Med ; 22(4): 474-483, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33073305

RESUMO

The CD137L-CD137 axis is a potent co-stimulatory immune checkpoint regulator that forms a bidirectional signaling pathway between the CD137 ligand (CD137L) and CD137 receptor to regulate immunological activities. This study investigated the potential involvement of the CD137L-CD137 axis on inflammasome-associated brain injury and neurological deficits in a mouse model of focal ischemic stroke. Cerebral ischemia was induced in male C57BL/6J wild-type (WT), CD137L-deficient (CD137L KO) and CD137-deficient (CD137 KO) mice by middle cerebral artery occlusion (MCAO; 60 min), followed by reperfusion (6 h and 24 h). Brain infarct volume and neurological deficit scores were significantly lower in both CD137L KO and CD137 KO mice compared to WT controls. Moreover, CD137L-deficient brains had significantly lower levels of the pyroptotic protein, NT-Gasdermin D, while CD137-deficient brains had significantly lower levels of the pro-apoptotic proteins, cleaved caspase-3, pyroptotic protein, NT-Gasdermin D, and of the secondary pyroptotic protein NT-Gasdermin E, following ischemic stroke. This protection by CD137L and CD137 deletion was associated with a significant decrease in inflammasome signaling. In conclusion, our data provide evidence for the first time that the CD137L-CD137 axis contributes to brain injury and neurological deficits by activating the inflammasome signaling pathway following ischemic stroke.


Assuntos
Ligante 4-1BB/fisiologia , Infarto da Artéria Cerebral Média/metabolismo , Inflamassomos/fisiologia , AVC Isquêmico/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/fisiologia , Ligante 4-1BB/deficiência , Alarminas/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Dano Encefálico Crônico/etiologia , Infarto Cerebral/etiologia , Infarto Cerebral/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , AVC Isquêmico/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Ligação a Fosfato/metabolismo , Receptores de Estrogênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/fisiologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/deficiência
12.
Brain Behav ; 10(1): e01444, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31804775

RESUMO

INTRODUCTION: Intermittent fasting (IF) has been suggested to have neuroprotective effects through the activation of multiple signaling pathways. Rodents fasted intermittently exhibit enhanced hippocampal neurogenesis and long-term potentiation (LTP) at hippocampal synapses compared with sedentary animals fed an ad libitum (AL) diet. However, the underlying mechanisms have not been studied. In this study, we evaluated the mechanistic gap in understanding IF-induced neurogenesis. METHODS: We evaluated the impact of 3 months of IF (12, 16, and 24 hr of food deprivation on a daily basis) on hippocampal neurogenesis in C57BL/6NTac mice using immunoblot analysis. RESULTS: Three-month IF significantly increased activation of the Notch signaling pathway (Notch 1, NICD1, and HES5), neurotrophic factor BDNF, and downstream cellular transcription factor, cAMP response element-binding protein (p-CREB). The expression of postsynaptic marker, PSD95, and neuronal stem cell marker, Nestin, was also increased in the hippocampus in response to 3-month IF. CONCLUSIONS: These findings suggest that IF may increase hippocampal neurogenesis involving the Notch 1 pathway.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Jejum/metabolismo , Hipocampo/metabolismo , Neurogênese/fisiologia , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Sinapses/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...