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
2.
Proc Natl Acad Sci U S A ; 120(25): e2300012120, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37307473

RESUMO

Aging compromises the repair and regrowth of brain vasculature and white matter during stroke recovery, but the underlying mechanisms remain elusive. To understand how aging jeopardizes brain tissue repair after stroke, we performed single-cell transcriptomic profiling of young adult and aged mouse brains at acute (3 d) and chronic (14 d) stages after ischemic injury, focusing a priori on the expression of angiogenesis- and oligodendrogenesis-related genes. We identified unique subsets of endothelial cells (ECs) and oligodendrocyte (OL) progenitors in proangiogenesis and pro-oligodendrogenesis phenotypic states 3 d after stroke in young mice. However, this early prorepair transcriptomic reprogramming was negligible in aged stroke mice, consistent with the impairment of angiogenesis and oligodendrogenesis observed during the chronic injury stages after ischemia. In the stroke brain, microglia and macrophages (MG/MΦ) may drive angiogenesis and oligodendrogenesis through a paracrine mechanism. However, this reparative cell-cell cross talk between MG/MΦ and ECs or OLs is impeded in aged brains. In support of these findings, permanent depletion of MG/MΦ via antagonism of the colony-stimulating factor 1 receptor resulted in remarkably poor neurological recovery and loss of poststroke angiogenesis and oligodendrogenesis. Finally, transplantation of MG/MΦ from young, but not aged, mouse brains into the cerebral cortices of aged stroke mice partially restored angiogenesis and oligodendrogenesis and rejuvenated sensorimotor function and spatial learning and memory. Together, these data reveal fundamental mechanisms underlying the age-related decay in brain repair and highlight MG/MΦ as effective targets for promoting stroke recovery.


Assuntos
Células Endoteliais , Acidente Vascular Cerebral , Animais , Camundongos , Encéfalo , Macrófagos , Análise de Sequência de RNA
3.
Stroke ; 54(4): 1088-1098, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36912142

RESUMO

BACKGROUND: Stroke is the primary cause of chronic disability in the elderly, as there are no neurorestorative treatments for those who do not qualify for recanalization therapy. Experimental evidence in stroke animals suggests that transplantation of bone marrow-derived human mesenchymal stem cells (hMSCs) holds promise, but hMSC transplantation has not been systematically tested in aged animals. We tested the hypothesis that poststroke hMSC transplantation improves stroke recovery in aged mice by promoting brain repair. METHODS: Permanent focal cerebral ischemia was induced in 20-month-old C57BL/6 male mice by distal middle cerebral artery occlusion. Bone marrow-derived hMSCs were expanded in vitro and then administrated intravenously into mice (1×106 cells in PBS) 24 hours after distal middle cerebral artery occlusion. Sensorimotor and cognitive functions, brain atrophy, and brain repair processes (neurogenesis, angiogenesis, oligodendrogenesis) were assessed for up to 56 days after stroke. RESULTS: Poststroke hMSC transplantation did not mitigate brain atrophy or improve neuronal survival at 56 days after distal middle cerebral artery occlusion. However, hMSC-treated mice displayed superior neurobehavioral performances in the open field, rotarod, adhesive removal, novel object, and Morris water maze tests compared with PBS-treated controls. hMSCs promoted white matter integrity and enhanced angiogenesis and oligodendrogenesis-but not neurogenesis-in the stroke brain. Positive correlations between neurobehavioral performance and brain repair profiles or white matter integrity were observed in stroke mice. CONCLUSIONS: Poststroke hMSC transplantation improves long-term stroke recovery in aged mice, likely via mechanisms involving enhanced microvascular regeneration and white matter restoration.


Assuntos
Isquemia Encefálica , Células-Tronco Mesenquimais , Acidente Vascular Cerebral , Camundongos , Humanos , Masculino , Animais , Idoso , Lactente , Infarto da Artéria Cerebral Média/cirurgia , Camundongos Endogâmicos C57BL , Encéfalo , Acidente Vascular Cerebral/terapia , Isquemia Encefálica/cirurgia , Modelos Animais de Doenças
4.
CNS Neurosci Ther ; 29(1): 129-139, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36341958

RESUMO

AIMS: This study aimed to investigate the effects of mild selective brain hypothermia on aged female ischemic mice. METHODS: A distal middle cerebral artery occlusion (dMCAO) model was established in aged female mice, who were then subjected to mild selective brain hypothermia immediately after the dMCAO procedure. Neurological behavioral examinations were conducted prior to and up to 35 days post-ischemia. Infarct volume, brain atrophy, pro-inflammation, and anti-inflammation microglia/macrophages phenotype and white matter injury were evaluated by immunofluorescence staining. Correlations between neurological behaviors and histological parameters were evaluated by Pearson product linear regression analysis. RESULTS: Sensorimotor and cognitive function tests confirmed the protective effect of mild selective brain hypothermia in elderly female ischemic mice. In addition, hypothermia decreased the infarct volume and brain atrophy induced by focal cerebral ischemia. Furthermore, hypothermia alleviated ischemia-induced short-term and long-term white matter injury, which was correlated with behavioral deficits. Finally, hypothermia suppressed the harmful immunological response by promoting the transformation of pro-inflammatory microglia/macrophages to anti-inflammatory phenotype. This polarization was negatively correlated with neuronal loss and white matter injury. CONCLUSION: Mild selective brain hypothermia promoted long-term functional recovery by alleviating white matter damage in an aged female mouse model of ischemia.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , Hipotermia Induzida , Hipotermia , Feminino , Camundongos , Animais , Encéfalo/patologia , Isquemia Encefálica/patologia , Microglia , Infarto da Artéria Cerebral Média/terapia , Infarto da Artéria Cerebral Média/patologia , Hipotermia Induzida/métodos , Anti-Inflamatórios/farmacologia
5.
J Clin Invest ; 132(15)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35912857

RESUMO

Immunomodulation holds therapeutic promise against brain injuries, but leveraging this approach requires a precise understanding of mechanisms. We report that CD8+CD122+CD49dlo T regulatory-like cells (CD8+ TRLs) are among the earliest lymphocytes to infiltrate mouse brains after ischemic stroke and temper inflammation; they also confer neuroprotection. TRL depletion worsened stroke outcomes, an effect reversed by CD8+ TRL reconstitution. The CXCR3/CXCL10 axis served as the brain-homing mechanism for CD8+ TRLs. Upon brain entry, CD8+ TRLs were reprogrammed to upregulate leukemia inhibitory factor (LIF) receptor, epidermal growth factor-like transforming growth factor (ETGF), and interleukin 10 (IL-10). LIF/LIF receptor interactions induced ETGF and IL-10 production in CD8+ TRLs. While IL-10 induction was important for the antiinflammatory effects of CD8+ TRLs, ETGF provided direct neuroprotection. Poststroke intravenous transfer of CD8+ TRLs reduced infarction, promoting long-term neurological recovery in young males or aged mice of both sexes. Thus, these unique CD8+ TRLs serve as early responders to rally defenses against stroke, offering fresh perspectives for clinical translation.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Animais , Linfócitos T CD8-Positivos/metabolismo , Feminino , Interleucina-10/genética , Interleucina-10/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo
6.
Prog Neurobiol ; 205: 102125, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34333040

RESUMO

Vascular cognitive impairment and dementia (VaD) is the second most common type of dementia caused by chronic vascular hypoperfusion. Adiponectin, one of the cytokines produced by adipocytes (adipocytokine), plays a role in CNS pathologies, but its specific function in VaD is unknown. Here, transcriptomic analyses on human brain tissues showed downregulation of adipocytokine/PPAR signaling in VaD patients, with prominent upregulation of pro-inflammatory responses. Using the murine asymmetric common carotid artery stenosis (ACAS) model, we discovered that the adiponectin/PPARγ axis is essential in reducing chronic hypoperfusion-induced cognitive deficits via modulation of microglial function. Adiponectin levels in the plasma increased early after VaD induction, but decreased in the cerebrospinal fluid in the late phase of VaD. Adiponectin deficiency worsened hippocampus-dependent cognitive deficits, exacerbated neuroinflammation and microglia/macrophage activation, and amplified neuronal loss, but these behavioral and histological outcomes were rescued by adipoRon, a small molecule agonist of the adiponectin receptors. AdipoRon boosted PPARγ expression and inhibited pro-inflammatory microglial responses in vitro, thereby protecting ischemic neurons in primary microglia-neuron cocultures. Microglia/macrophage-specific knockout of PPARγ abolished the neuroprotective effects of adipoRon. Collectively, these data confirm the importance of adiponectin/PPARγ signaling in maintaining cognitive functions in chronic hypoperfusion-induced dementia, and thus provide novel therapeutic targets for VaD.


Assuntos
Disfunção Cognitiva , Adiponectina , Animais , Cognição , Demência , Humanos , Camundongos , Microglia , Doenças Neuroinflamatórias , Fármacos Neuroprotetores , PPAR gama
7.
CNS Neurosci Ther ; 27(5): 515-527, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33650313

RESUMO

Stroke is a leading cause of disability and mortality, with limited treatment options. After stroke injury, microglia and CNS-resident macrophages are rapidly activated and regulate neuropathological processes to steer the course of functional recovery. To accelerate this recovery, microglia can engulf dying cells and clear irreparably-damaged tissues, thereby creating a microenvironment that is more suitable for the formation of new neural circuitry. In addition, monocyte-derived macrophages cross the compromised blood-brain barrier to infiltrate the injured brain. The specific functions of myeloid lineage cells in brain injury and repair are diverse and dependent on phenotypic polarization statuses. However, it remains to be determined to what degree the CNS-invading macrophages occupy different functional niches from CNS-resident microglia. In this review, we describe the physiological characteristics and functions of microglia in the developing and adult brain. We also review (a) the activation and phenotypic polarization of microglia and macrophages after stroke, (b) molecular mechanisms that control polarization status, and (c) the contribution of microglia to brain pathology versus repair. Finally, we summarize current breakthroughs in therapeutic strategies that calibrate microglia/macrophage responses after stroke.


Assuntos
Lesões Encefálicas/patologia , Macrófagos/patologia , Microglia/patologia , Acidente Vascular Cerebral/patologia , Adulto , Animais , Lesões Encefálicas/etiologia , Polaridade Celular , Humanos , Ativação de Macrófagos , Acidente Vascular Cerebral/complicações
8.
Transl Stroke Res ; 12(3): 474-495, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33128703

RESUMO

As an integral part of the innate immune system of the brain, resident microglia must react rapidly to the onset of brain injury and neurological disease. These dynamic cells then continue to shift their phenotype along a multidimensional continuum with overlapping pro- and anti-inflammatory states, allowing them to adapt to microenvironmental changes during the progression of brain disorders. However, the ability of microglia to shift phenotype through nimble molecular, structural, and functional changes comes at a cost, as the extreme pro-inflammatory states may prevent these professional phagocytes from clearing toxic debris and secreting tissue-repairing neurotrophic factors. Evolution has strongly favored heterogeneity in microglia in both the spatial and temporal dimensions-they can assume diverse roles in different brain regions, throughout the course of brain development and aging, and during the spatiotemporal progression of brain injuries and neurological diseases. Age and sex differences add further diversity to microglia functional status under physiological and pathological conditions. This article reviews recent advances in our knowledge of microglia with emphases on molecular mediators of phenotype shifts and functional diversity. We describe microglia-targeted therapeutic opportunities, including pharmacologic modulation of phenotype and repopulation of the brain with fresh microglia. With the advent of powerful new tools, research on microglia has recently accelerated in pace and may translate into potential therapeutics against brain injury and neurological disease.


Assuntos
Lesões Encefálicas , Doenças do Sistema Nervoso , Envelhecimento , Encéfalo , Feminino , Humanos , Masculino , Microglia
9.
Chronic Dis Transl Med ; 6(1): 46-54, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32226934

RESUMO

BACKGROUND: Regulated upon activation, normal T-cell expressed, and secreted (RANTES) is a chemokine actively involved in the initiation and progression of atherosclerosis (AS), which is the major cause of ischemic cerebrovascular disease (ICVD). This study aimed to determine the associations between circulating RANTES level and overall AS conditions of cardiac and cerebral vessel beds in patients with ICVD. METHODS: Patients with ICVD admitted to the department of neurology of Xuanwu Hospital from April 1, 2019 to June 30, 2019 were prospectively enrolled in the study. Plasma RANTES level was measured by enzyme-linked immunosorbent assay to represent the circulating RANTES level. The integrated AS burden of the cervicocephalic and coronary arteries was examined using computed tomography angiography and reflected by "cardio-cerebral AS burden (CCAB)" as a continuous variable. Then, the relationship of plasma RANTES level and CCAB in patients with ICVD was analyzed by correlation analyses and general linear models. RESULTS: A total of 40 patients with ICVD were included in the study. There was a significant positive correlation between CCAB and plasma RANTES level in ICVD (r = 0.786, P < 0.001), independent of age, sex, acute or chronic phase of ICVD, and mono or dual antiplatelet therapy (adjusted B for ln RANTES, 12.063; 95% confidence interval, 7.572-16.533). The association of plasma RANTES level with AS conditions (burden, severity, and extent) in single cardiac or cerebral vessel bed was similar to that with CCAB, but the correlation coefficient for CCAB was higher (increment ranged from 0.126 to 0.397). CONCLUSIONS: Plasma RANTES level was an independent indicator for the integrated AS burden of the cervicocephalic and coronary arteries in ICVD. Comprehensive evaluation of AS conditions using the novel continuous index CCAB might be important in revealing the systematic relationship between circulating RANTES and AS in patients with ICVD.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA