Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.180
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Nature ; 568(7752): 336-343, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30996318

RESUMO

The brains of humans and other mammals are highly vulnerable to interruptions in blood flow and decreases in oxygen levels. Here we describe the restoration and maintenance of microcirculation and molecular and cellular functions of the intact pig brain under ex vivo normothermic conditions up to four hours post-mortem. We have developed an extracorporeal pulsatile-perfusion system and a haemoglobin-based, acellular, non-coagulative, echogenic, and cytoprotective perfusate that promotes recovery from anoxia, reduces reperfusion injury, prevents oedema, and metabolically supports the energy requirements of the brain. With this system, we observed preservation of cytoarchitecture; attenuation of cell death; and restoration of vascular dilatory and glial inflammatory responses, spontaneous synaptic activity, and active cerebral metabolism in the absence of global electrocorticographic activity. These findings demonstrate that under appropriate conditions the isolated, intact large mammalian brain possesses an underappreciated capacity for restoration of microcirculation and molecular and cellular activity after a prolonged post-mortem interval.


Assuntos
Autopsia , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Circulação Cerebrovascular , Microcirculação , Suínos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Caspase 3/metabolismo , Sobrevivência Celular , Artérias Cerebrais/fisiologia , Modelos Animais de Doenças , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Inflamação/metabolismo , Inflamação/patologia , Neuroglia/citologia , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia , Perfusão , Traumatismo por Reperfusão/prevenção & controle , Suínos/sangue , Sinapses/metabolismo , Sinapses/patologia , Fatores de Tempo , Vasodilatação
2.
Cell Mol Neurobiol ; 44(1): 58, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39105862

RESUMO

This article comprehensively reviews how cerebral hypoxia impacts the physiological state of neurons and dendritic spines through a series of molecular changes, and explores the causal relationship between these changes and neuronal functional impairment. As a severe pathological condition, cerebral hypoxia can significantly alter the morphology and function of neurons and dendritic spines. Specifically, dendritic spines, being the critical structures for neurons to receive information, undergo changes such as a reduction in number and morphological abnormalities under hypoxic conditions. These alterations further affect synaptic function, leading to neurotransmission disorders. This article delves into the roles of molecular pathways like MAPK, AMPA receptors, NMDA receptors, and BDNF in the hypoxia-induced changes in neurons and dendritic spines, and outlines current treatment strategies. Neurons are particularly sensitive to cerebral hypoxia, with their apical dendrites being vulnerable to damage, thereby affecting cognitive function. Additionally, astrocytes and microglia play an indispensable role in protecting neuronal and synaptic structures, regulating their normal functions, and contributing to the repair process following injury. These studies not only contribute to understanding the pathogenesis of related neurological diseases but also provide important insights for developing novel therapeutic strategies. Future research should further focus on the dynamic changes in neurons and dendritic spines under hypoxic conditions and their intrinsic connections with cognitive function.


Assuntos
Espinhas Dendríticas , Neurônios , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Animais , Humanos , Neurônios/metabolismo , Neurônios/patologia , Hipóxia Encefálica/patologia , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/fisiopatologia
3.
Exp Cell Res ; 391(1): 111988, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32251645

RESUMO

Tripartite motif 16 (TRIM16) has emerged as a novel oxidative stress-responsive protein that confers cytoprotective effects by reinforcing the cellular antioxidant system. However, whether TRIM16 is involved in regulating oxidative stress during cerebral ischemia/reperfusion injury remains unclear. In the present study, we aimed to explore the potential function and molecular mechanism of TRIM16 in regulating oxidative stress in neurons induced by oxygen-glucose deprivation/reoxygenation (OGD/R) in vitro. Here, we found that OGD/R exposure resulted in a significant induction of TRIM16 expression in neurons. Depletion of TRIM16 by siRNA-mediated gene knockdown markedly upregulated the sensitivity of neurons to OGD/R-induced apoptosis and reactive oxygen species (ROS) generation. Notably, upregulation of TRIM16 expression significantly alleviated OGD/R-induced apoptosis and ROS generation in neurons. Moreover, TRIM16 overexpression markedly increased nuclear factor erythroid 2-related factor 2 (Nrf2) expression and enhanced Nrf2/antioxidant response element (ARE) activation associated with downregulation of kelch-like ECH-associated protein 1 (Keap1) expression. Restoration of Keap1 significantly reversed the TRIM16-mediated promotion effect on Nrf2/ARE activation. In addition, knockdown of Nrf2 also markedly abrogated the TRIM16-conferred neuroprotective effect in OGD/R-exposed neurons. Taken together, our results of our study demonstrate that induction of TRIM16 confers a cytoprotective effect in OGD/R-exposed neurons through enhancement of Nrf2/ARE antioxidant signaling via downregulation of Keap1. These findings suggest that TRIM16 may play a critical role in cerebral ischemia/reperfusion injury and serve as a promising target for neuroprotection.


Assuntos
Antioxidantes/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/biossíntese , Fator 2 Relacionado a NF-E2/metabolismo , Neuroproteção/fisiologia , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Anaerobiose , Animais , Apoptose/fisiologia , Linhagem Celular , Glucose/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Hipóxia Encefálica/patologia , Camundongos , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética
4.
J Stroke Cerebrovasc Dis ; 30(6): 105739, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33765634

RESUMO

OBJECTIVES: Endovascular treatment (EVT) has become the standard of care for acute ischemic stroke. Despite successful recanalization, a limited subset of patients benefits from the new treatment. Human MRI studies have shown that during removal of the thrombus, a shower of microclots is released from the initial thrombus, possibly causing new ischemic lesions. The aim of the current study is to quantify tissue damage following microembolism. MATERIALS AND METHODS: In a rat model, microembolism was generated by injection of a mixture of polystyrene fluorescent microspheres (15, 25 and 50 µm in diameter). The animals were killed at three time-points: day 1, 3 or 7. AMIRA and IMARIS software was used for 3D reconstruction of brain structure and damage, respectively. CONCLUSIONS: Microembolism induces ischemia, hypoxia and infarction. Infarcted areas persist, but hypoxic regions recover over time suggesting that repair processes in the brain rescue the regions at risk.


Assuntos
Infarto Encefálico/etiologia , Isquemia Encefálica/etiologia , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular , Hipóxia Encefálica/etiologia , Embolia Intracraniana/complicações , Oxigênio/sangue , Animais , Infarto Encefálico/sangue , Infarto Encefálico/patologia , Infarto Encefálico/fisiopatologia , Isquemia Encefálica/sangue , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Modelos Animais de Doenças , Feminino , Hipóxia Encefálica/sangue , Hipóxia Encefálica/patologia , Hipóxia Encefálica/fisiopatologia , Embolia Intracraniana/sangue , Embolia Intracraniana/patologia , Embolia Intracraniana/fisiopatologia , Masculino , Ratos Wistar , Recuperação de Função Fisiológica , Fatores de Tempo
5.
J Neurosci ; 39(7): 1320-1331, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30587544

RESUMO

Individuals with sleep apnea often exhibit changes in cognitive behaviors consistent with alterations in the hippocampus. It is hypothesized that adult neurogenesis in the dentate gyrus is an ongoing process that maintains normal hippocampal function in many mammalian species, including humans. However, the impact of chronic intermittent hypoxia (IH), a principal consequence of sleep apnea, on hippocampal adult neurogenesis remains unclear. Using a murine model, we examined the impact of 30 d of IH (IH30) on adult neurogenesis and synaptic plasticity in the dentate gyrus. Although IH30 did not affect paired-pulse facilitation, IH30 suppressed long-term potentiation (LTP). Immunohistochemical experiments also indicate that IH perturbs multiple aspects of adult neurogenesis. IH30 increased the number of proliferating Sox2+ neural progenitor cells in the subgranular zone yet reduced the number of doublecortin-positive neurons. Consistent with these findings, cell lineage tracing revealed that IH30 increased the proportion of radial glial cells in the subgranular zone, yet decreased the proportion of adult-born neurons in the dentate gyrus. While administration of a superoxide anion scavenger during IH did not prevent neural progenitor cell proliferation, it mitigated the IH-dependent suppression of LTP and prevented adult-born neuron loss. These data demonstrate that IH causes both reactive oxygen species-dependent and reactive oxygen species-independent effects on adult neurogenesis and synaptic plasticity in the dentate gyrus. Our findings identify cellular and neurophysiological changes in the hippocampus that may contribute to cognitive and behavioral deficits occurring in sleep apnea.SIGNIFICANCE STATEMENT Individuals with sleep apnea experience periods of intermittent hypoxia (IH) that can negatively impact many aspects of brain function. Neurons are continually generated throughout adulthood to support hippocampal physiology and behavior. This study demonstrates that IH exposure attenuates hippocampal long-term potentiation and reduces adult neurogenesis. Antioxidant treatment mitigates these effects indicating that oxidative signaling caused by IH is a significant factor that impairs synaptic plasticity and reduces adult neurogenesis in the hippocampus.


Assuntos
Giro Denteado/patologia , Hipóxia Encefálica/patologia , Neurogênese , Plasticidade Neuronal , Animais , Linhagem da Célula , Proliferação de Células , Proteínas do Domínio Duplacortina , Potenciais Pós-Sinápticos Excitadores , Feminino , Sequestradores de Radicais Livres/farmacologia , Hipóxia Encefálica/etiologia , Potenciação de Longa Duração , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Neurais/patologia , Neuroglia/patologia , Neuropeptídeos/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/genética , Síndromes da Apneia do Sono/complicações , Síndromes da Apneia do Sono/fisiopatologia
6.
Cell Physiol Biochem ; 54(6): 1231-1248, 2020 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-33326735

RESUMO

BACKGROUND/AIMS: Obstructive sleep apnea (OSA) is characterized by repeated episodes of complete or partial obstruction of the upper airways, leading to chronic intermittent hypoxia (IH). OSA patients are considered at high cerebrovascular risk and may also present cognitive impairment. One hypothesis explored is that disturbances may be linked to blood-brain barrier (BBB) dysfunction. The BBB is a protective barrier separating the brain from blood flow. The BBB limits the paracellular pathway through tight and adherens junctions, and the transcellular passage by efflux pumps (ABC transporters). The aims of this study were to evaluate the impact of IH and sustained hypoxia (SH) on a validated in vitro BBB model and to investigate the factors expressed under both conditions. METHODS: Exposure of endothelial cells (HBEC-5i) in our in vitro model of BBB to hypoxia was performed using IH cycles: 1% O2-35 min/18% O2-25 min for 6 cycles or 6 h of SH at 1% O2. After exposure, we studied the cytotoxicity and the level of ROS in our cells. We measured the apparent BBB permeability using sodium fluorescein, FITC-dextran and TEER measurement. Whole cell ELISA were performed to evaluate the expression of tight junctions, ABC transporters, HIF-1α and Nrf2. The functionality of ABC transporters was evaluated with accumulation studies. Immunofluorescence assays were also conducted to illustrate the whole cell ELISAs. RESULTS: Our study showed that 6 h of IH or SH induced a BBB disruption marked by a significant decrease in junction protein expressions (claudin-5, VE-cadherin, ZO-1) and an increase in permeability. We also observed an upregulation in P-gp protein expression and functionality and a downregulation in BCRP. Hypoxia induced production of ROS, Nrf2 and HIF-1α. They were expressed in both sustained and intermittent conditions, but the expression and the activity of P-gp and BCRP were different. CONCLUSION: Understanding these mechanisms seems essential in order to propose new therapeutic strategies for patients with OSA.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/biossíntese , Barreira Hematoencefálica/metabolismo , Regulação da Expressão Gênica , Hipóxia Encefálica/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Modelos Cardiovasculares , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas de Neoplasias/biossíntese , Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Barreira Hematoencefálica/patologia , Linhagem Celular , Humanos , Hipóxia Encefálica/patologia
7.
J Surg Res ; 246: 6-18, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31541709

RESUMO

BACKGROUND: Remote ischemic postconditioning (RIPost) has been shown to reduce the ischemia-reperfusion injury of the heart and brain. However, the protection mechanisms have not yet been fully elucidated. We have observed that RIPost could alleviate the brain injury after cardiac arrest (CA). The aim of this study was to explore whether α7 nicotinic acetylcholine receptor (α7nAChR) mediates the neuroprotection of RIPost in a rat model of asphyxial CA. MATERIALS AND METHODS: Asphyxial CA model was induced by occlusion of the tracheal tube for 8 min and resuscitated later. RIPost produced by three cycles of 15-min occlusion and 15-min release of the right hind limb by a tourniquet was performed respectively at the moment and the third hour after restoration of spontaneous circulation. The α7nAChR agonist PHA-543613 and the antagonist methyllycaconitine (MLA) were used to investigate the role of α7nAChR in mediating neuroprotective effects. RESULTS: Results showed that α7nAChR was decreased in hippocampus and cortex after resuscitation, whereas RIPost could attenuate the reduction. The use of PHA-543613 provided neuroprotective effects against cerebral injury after CA. Furthermore, RIPost decreased the levels of neuron-specific enolase, inflammatory mediators, the number of apoptotic cells, and phosphorylation of nuclear factor-κB while increased the phosphorylation of signal transducer and activator of transcription-3. However, the above effects of RIPost were attenuated by α7nAChR antagonist methyllycaconitine. CONCLUSIONS: Neuroprotection of RIPost was related with the activation of α7nAChR, which could suppress nuclear factor-κB and activate signal transducer and activator of transcription-3 in a rat asphyxial CA model.


Assuntos
Parada Cardíaca/terapia , Hipóxia Encefálica/terapia , Pós-Condicionamento Isquêmico , Neuroproteção/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Aconitina/análogos & derivados , Aconitina/farmacologia , Animais , Asfixia/complicações , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Modelos Animais de Doenças , Parada Cardíaca/etiologia , Hipocampo/irrigação sanguínea , Hipocampo/patologia , Humanos , Hipóxia Encefálica/etiologia , Hipóxia Encefálica/patologia , Masculino , NF-kappa B/metabolismo , Neuroproteção/efeitos dos fármacos , Quinuclidinas/farmacologia , Ratos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Resultado do Tratamento , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores
8.
Int J Mol Sci ; 21(11)2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32492921

RESUMO

Multimodal continuous bedside monitoring is increasingly recognized as a promising option for early treatment stratification in patients at risk for ischemia during neurocritical care. Modalities used at present are, for example, oxygen availability and subdural electrocorticography. The assessment of mitochondrial function could be an interesting complement to these modalities. For instance, flavin adenine dinucleotide (FAD) fluorescence permits direct insight into the mitochondrial redox state. Therefore, we explored the possibility of using FAD fluorometry to monitor consequences of hypoxia in brain tissue in vitro and in vivo. By combining experimental results with computational modeling, we identified the potential source responsible for the fluorescence signal and gained insight into the hypoxia-associated metabolic changes in neuronal energy metabolism. In vitro, hypoxia was characterized by a reductive shift of FAD, impairment of synaptic transmission and increasing interstitial potassium [K+]o. Computer simulations predicted FAD changes to originate from the citric acid cycle enzyme α-ketoglutarate dehydrogenase and pyruvate dehydrogenase. In vivo, the FAD signal during early hypoxia displayed a reductive shift followed by a short oxidation associated with terminal spreading depolarization. In silico, initial tissue hypoxia followed by a transient re-oxygenation phase due to glucose depletion might explain FAD dynamics in vivo. Our work suggests that FAD fluorescence could be readily used to monitor mitochondrial function during hypoxia and represents a potential diagnostic tool to differentiate underlying metabolic processes for complementation of multimodal brain monitoring.


Assuntos
Encéfalo/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Fluorescência , Hipóxia Encefálica/metabolismo , Mitocôndrias/metabolismo , Animais , Encéfalo/fisiopatologia , Ciclo do Ácido Cítrico , Simulação por Computador , Metabolismo Energético , Fluorometria , Hipóxia Encefálica/patologia , Masculino , Mitocôndrias/patologia , Oxirredução , Oxigênio/metabolismo , Potássio/metabolismo , Ratos , Ratos Wistar
9.
J Neurosci ; 38(5): 1100-1113, 2018 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-29246927

RESUMO

Many Preterm-born children suffer from neurobehavioral disorders. Premature birth terminates the hypoxic in utero environment and supply of maternal hormones. As the production of interneurons continues until the end of pregnancy, we hypothesized that premature birth would disrupt interneuron production and that restoration of the hypoxic milieu or estrogen treatment might reverse interneuron generation. To test these hypotheses, we compared interneuronal progenitors in the medial ganglionic eminences (MGEs), lateral ganglionic eminences (LGEs), and caudal ganglionic eminences (CGEs) between preterm-born [born on embryonic day (E) 29; examined on postnatal day (D) 3 and D7] and term-born (born on E32; examined on D0 and D4) rabbits at equivalent postconceptional ages. We found that both total and cycling Nkx2.1+, Dlx2+, and Sox2+ cells were more abundant in the MGEs of preterm rabbits at D3 compared with term rabbits at D0, but not in D7 preterm relative to D4 term pups. Total Nkx2.1+ progenitors were also more numerous in the LGEs of preterm pups at D3 compared with term rabbits at D0. Dlx2+ cells in CGEs were comparable between preterm and term pups. Simulation of hypoxia by dimethyloxalylglycine treatment did not affect the number of interneuronal progenitors. However, estrogen treatment reduced the density of total and proliferating Nkx2.1+ and Dlx2+ cells in the MGEs and enhanced Ascl1 transcription factor. Estrogen treatment also reduced Ki67, c-Myc, and phosphorylation of retinoblastoma protein, suggesting inhibition of the G1-to-S phase transition. Hence, preterm birth disrupts interneuron neurogenesis in the MGE and estrogen treatment reverses interneuron neurogenesis in preterm newborns by cell-cycle inhibition and elevation of Ascl1. We speculate that estrogen replacement might partially restore neurogenesis in human premature infants.SIGNIFICANCE STATEMENT Prematurity results in developmental delays and neurobehavioral disorders, which might be ascribed to disturbances in the development of cortical interneurons. Here, we show that preterm birth disrupts interneuron neurogenesis in the medial ganglionic eminence (MGE) and, more importantly, that estrogen treatment reverses this perturbation in the population of interneuron progenitors in the MGE. The estrogen seems to restore neurogenesis by inhibiting the cell cycle and elevating Ascl1 expression. As preterm birth causes plasma estrogen level to drop 100-fold, the estrogen replacement in preterm infants is physiological. We speculate that estrogen replacement might ameliorate disruption in production of interneurons in human premature infants.


Assuntos
Animais Recém-Nascidos/fisiologia , Estrogênios/uso terapêutico , Interneurônios/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Animais , Feminino , Gânglios/citologia , Gânglios/crescimento & desenvolvimento , Gânglios/metabolismo , Proteínas de Homeodomínio/metabolismo , Hipóxia Encefálica/induzido quimicamente , Hipóxia Encefálica/patologia , Antígeno Ki-67/metabolismo , Células-Tronco Neurais/metabolismo , Gravidez , Nascimento Prematuro , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , Coelhos , Fatores de Transcrição SOXB1/metabolismo , Fator Nuclear 1 de Tireoide/metabolismo
10.
Neurochem Res ; 44(6): 1425-1436, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30448928

RESUMO

Post-conditioning is exposure of an injured organism to the same harmful factors but of milder intensity which mobilizes endogenous protective mechanisms. Recently, we have developed a novel noninvasive post-conditioning (PostC) protocol involving three sequential episodes of mild hypobaric hypoxia which exerts pronounced neuroprotective action. In particular, it prevents development of pathological cascades caused by severe hypobaric hypoxia (SH) such as cellular loss, lipid peroxidation, abnormal neuroendocrine responses and behavioural deficit in experimental animals. Development of these post-hypoxic pathological effects has been associated with the delayed reduction of hypoxia-inducible factor 1 (HIF1) regulatory α-subunit levels in rat hippocampus, whereas PostC up-regulated it. The present study has been aimed at experimental examination of the hypothesis that intrinsic mechanisms underlying the neuroprotective and antioxidant effects of PostC involves HIF1-dependent stimulation of the pentose phosphate pathway (PPP). We have observed that SH leads to a decrease of glucose-6-phosphate dehydrogenase (G6PD) activity in the hippocampus and neocortex of rats as well as to a reduction in NADPH and total glutathione levels. This depletion of the antioxidant defense system together with excessive lipid peroxidation during the reoxygenation phase resulted in increased oxidative stress and massive cellular death observed after SH. In contrast, PostC led to normalization of G6PD activity, stabilization of the NADPH and total glutathione levels and thereby resulted in recovery of the cellular redox state and prevention of neuronal death. Our data suggest that stabilization of the antioxidant system via HIF1-associated PPP regulation represents an important neuroprotective mechanism enabled by PostC.


Assuntos
Encéfalo/metabolismo , Hipóxia Encefálica/prevenção & controle , Hipóxia Encefálica/terapia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Neuroproteção/fisiologia , Via de Pentose Fosfato/fisiologia , Animais , Encéfalo/patologia , Glucosefosfato Desidrogenase/metabolismo , Glutationa/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Masculino , NADP/metabolismo , Neocórtex/metabolismo , Neocórtex/patologia , Estresse Oxidativo/fisiologia , Ratos Wistar
11.
Pediatr Res ; 85(5): 591-595, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30666047

RESUMO

The assessment of the wellbeing of the cardiovascular status in premature infants has come to the forefront in recent years. There is an increasing realisation that myocardial performance, systemic blood flow and end-organ perfusion (particularly during the transitional period) play an important role in determining short and long-term outcomes in this population. The recent open access series on Neonatologist Performed Echocardiography (NPE) published in this journal outline the necessary techniques for image acquisition and analysis and provide a framework for the potential clinical applications of NPE in neonatal, and specifically preterm care. In this "Future Perspectives" review, we describe the important determinants of adequate cellular metabolism and myocardial performance (e.g. loading conditions, intrinsic contractility and morphological change), we discuss the maladaptive state of the preterm cardiovascular system, and highlight the emerging role that non-invasive echocardiography techniques, such as deformation analysis, serve in identifying the underlying physiological basis for cardiovascular instability.


Assuntos
Ecocardiografia/métodos , Coração/fisiopatologia , Doenças do Prematuro/diagnóstico por imagem , Contração Miocárdica/fisiologia , Miocárdio/patologia , Pressão Sanguínea , Débito Cardíaco , Diástole , Elasticidade , Idade Gestacional , Coração/diagnóstico por imagem , Humanos , Hipóxia Encefálica/diagnóstico por imagem , Hipóxia Encefálica/patologia , Hipóxia Encefálica/fisiopatologia , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/patologia , Doenças do Prematuro/fisiopatologia , Miócitos Cardíacos/metabolismo , Sístole , Resistência Vascular
12.
J Stroke Cerebrovasc Dis ; 28(3): 595-603, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30482485

RESUMO

BACKGROUND: Survival following cardiac arrest (CA) and subsequent cardiopulmonary resuscitation (CPR), to a great extent, depends on brain damage. Adipose-derived stem cells (ADSCs), as a source of paracrine growth factors and the capacity of neural differentiation may reduce this brain damage. OBJECTIVE: The purpose of this study is to evaluate the protection of ADSCs to brain damage following CPR. METHODS: Rats were divided into 3 groups, sham, CA, and ADSCs group. Rats in sham group went through sham surgery. Rats in CA group went through CA, CPR, and injection PBS (phosphate buffer saline). Rats in ADSCs group went through CA, CPR, and intravenous injection of ADSCs. Rats in sham group were sacrificed immediately after operation. At 24, 72, and 168 hours after return of spontaneous circulation operation, rats in CA and ADSCs group were randomly selected and sacrificed. Brain damage was evaluated by using Neurological Deficit Scale (NDS) score, hippocampal pathology, serum level of S100ß, and apoptosis ratio of hippocampal neurons. Protein of brain derived neurotrophic factor (BDNF) and IL-6 (interleukin-6) in the hippocampus were detected. RESULTS: Compared with sham group, CA and ADSCs group showed a decrease in NDS score, an increased apoptosis ratio of hippocampal nerve cells, increased serum level of S100-ß, and a significant increase in neuroprotective IL-6 and BDNF. In comparison to CA group, ADSCs group had a mild degree of brain damage and higher expression of IL-6 and BDNF. CONCLUSIONS: In the acute stage of cerebral injury following CA, ADSCs might improve the prognosis of brain damage by stimulating the expression of neuroprotective IL-6 and BDNF.


Assuntos
Tecido Adiposo/transplante , Parada Cardíaca/complicações , Hipocampo/patologia , Hipóxia Encefálica/prevenção & controle , Neurônios/patologia , Transplante de Células-Tronco , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Apoptose , Comportamento Animal , Biomarcadores/sangue , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Reanimação Cardiopulmonar , Células Cultivadas , Modelos Animais de Doenças , Parada Cardíaca/fisiopatologia , Parada Cardíaca/terapia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Hipóxia Encefálica/etiologia , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Interleucina-6/metabolismo , Masculino , Neurônios/metabolismo , Comunicação Parácrina , Ratos Sprague-Dawley , Subunidade beta da Proteína Ligante de Cálcio S100/sangue , Transdução de Sinais
13.
J Cell Mol Med ; 22(6): 3167-3182, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29536658

RESUMO

Hypoxia-ischaemia (HI) remains a major cause of foetal brain damage presented a scarcity of effective therapeutic approaches. Dexmedetomidine (DEX) and microRNA-140-5p (miR-140-5p) have been highlighted due to its potentially significant role in the treatment of cerebral ischaemia. This study was to investigate the role by which miR-140-5p provides cerebral protection using DEX to treat hypoxic-ischaemic brain damage (HIBD) in neonatal rats via the Wnt/ß-catenin signalling pathway. The HIBD rat models were established and allocated into various groups with different treatment plans, and eight SD rats into sham group. The learning and memory ability of the rats was assessed. Apoptosis and pathological changes in the hippocampus CA1 region and expressions of the related genes of the Wnt/ß-catenin signalling pathway as well as the genes responsible of apoptosis were detected. Compared with the sham group, the parameters of weight, length growth, weight ratio between hemispheres, the rate of reaching standard, as well as Bcl-2 expressions, were all increased. Furthermore, observations of increased levels of cerebral infarction volume, total mortality rate, response times, total response duration, expressions of Wnt1, ß-catenin, TCF-4, E-cadherin, apoptosis rate of neurons, and Bax expression were elevated. Following DEX treatment, the symptoms exhibited by HIBD rats were ameliorated. miR-140-5p and si-Wnt1 were noted to attenuate the progression of HIBD. Our study demonstrates that miR-140-5p promotes the cerebral protective effects of DEX against HIBD in neonatal rats by targeting the Wnt1 gene through via the negative regulation of the Wnt/ß-catenin signalling pathway.


Assuntos
Dexmedetomidina/administração & dosagem , Hipóxia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , MicroRNAs/genética , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hipóxia Encefálica/genética , Hipóxia Encefálica/patologia , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos , Via de Sinalização Wnt , Proteína Wnt1/genética , beta Catenina/genética
14.
Neurobiol Dis ; 116: 39-52, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29723606

RESUMO

Chronic hypoxic stress results in deposition of lipofuscin granules in the CA3 region of hippocampal neurons which contributes to neurodegeneration and accelerated neuronal aging. Oxidative stress and mitophagy during hypoxia are crucial to cause aggregation of these lipofuscin granules in hypoxic neurons. Salidroside, a glucoside derivative of ß-Tyrosol, has been reported to protect hypoxic neurons through maintenance of mitochondrial activity. The present study is aimed at investigating the potential of Salidroside in preventing mitophagy during chronic hypoxia and identification of the molecular targets and underlying signaling mechanisms. In-silico analysis for interaction of salidroside with Bcl-xL was carried out using VLife MDS software. The prophylactic efficacy of Salidroside for amelioration of global hypoxia induced neuronal aging was studied in adult male Sprague-Dawley rats exposed to hypobaric hypoxia simulating an altitude of 7600 m for 21 days. Salidroside was supplemented at a daily dose of 25 mg kg-1b.w. p.o. during hypoxic exposure. Ultra-structural and immune-histological studies were conducted to study lipofuscin aggregation and mitophagy. In-silico findings on salidroside mediated stabilization of Bcl-xL were validated by investigating its effect on downstream signaling molecules involved in mitophagy. Administration of Salidroside reduced deposition of lipofuscin in hypoxic CA3 hippocampal neurons and prevented mitophagy. Salidroside stabilizes Bcl-xL in hypoxic neurons resulting in inhibition of PGAM5 phosphatase activity and maintenance of FUNDC1 in phosphorylated state. Salidroside mediated inhibition of pFUNDC1 dephosphorylation prevents FUNDC1-LC3 II interaction which is crucial for mitophagy. The present study demonstrates potential of Salidroside in preventing lipofuscin deposition during chronic hypoxic stress.


Assuntos
Região CA3 Hipocampal/metabolismo , Glucosídeos/metabolismo , Hipóxia Encefálica/metabolismo , Mitofagia/fisiologia , Neurônios/metabolismo , Fenóis/metabolismo , Proteína bcl-X/metabolismo , Animais , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/ultraestrutura , Glucosídeos/farmacologia , Hipóxia Encefálica/patologia , Masculino , Mitofagia/efeitos dos fármacos , Simulação de Acoplamento Molecular/métodos , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Fenóis/farmacologia , Estrutura Secundária de Proteína , Ratos , Ratos Sprague-Dawley , Proteína bcl-X/química
15.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R611-R622, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351418

RESUMO

Moderate anemia is associated with increased mortality and morbidity, including acute kidney injury (AKI), in surgical patients. A red blood cell (RBC)-specific antibody model was utilized to determine whether moderate subacute anemia could result in tissue hypoxia as a potential mechanism of injury. Cardiovascular and hypoxic cellular responses were measured in transgenic mice capable of expressing hypoxia-inducible factor-1α (HIF-1α)/luciferase activity in vivo. Antibody-mediated anemia was associated with mild intravascular hemolysis (6 h) and splenic RBC sequestration ( day 4), resulting in a nadir hemoglobin concentration of 89 ± 13 g/l on day 4. At this time point, renal tissue oxygen tension (PtO2) was decreased in anemic mice relative to controls (13.1 ± 4.3 vs. 20.8 ± 3.7 mmHg, P < 0.001). Renal tissue hypoxia was associated with an increase in HIF/luciferase expression in vivo ( P = 0.04) and a 20-fold relative increase in renal erythropoietin mRNA transcription ( P < 0.001) but no increase in renal blood flow ( P = 0.67). By contrast, brain PtO2 was maintained in anemic mice relative to controls (22.7 ± 5.2 vs. 23.4 ± 9.8 mmHg, P = 0.59) in part because of an increase in internal carotid artery blood flow (80%, P < 0.001) and preserved cerebrovascular reactivity. Despite these adaptive changes, an increase in brain HIF-dependent mRNA levels was observed (erythropoietin: P < 0.001; heme oxygenase-1: P = 0.01), providing evidence for subtle cerebral tissue hypoxia in anemic mice. These data demonstrate that moderate subacute anemia causes significant renal tissue hypoxia, whereas adaptive cerebrovascular responses limit the degree of cerebral tissue hypoxia. Further studies are required to assess whether hypoxia is a mechanism for acute kidney injury associated with anemia.


Assuntos
Injúria Renal Aguda/sangue , Anemia/sangue , Anticorpos Monoclonais , Encéfalo/irrigação sanguínea , Eritrócitos/metabolismo , Hipóxia Encefálica/sangue , Rim/irrigação sanguínea , Oxigênio/sangue , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Anemia/imunologia , Anemia/patologia , Anemia/fisiopatologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Circulação Cerebrovascular , Modelos Animais de Doenças , Eritrócitos/imunologia , Eritrócitos/patologia , Eritropoetina/genética , Eritropoetina/metabolismo , Glicoforinas/sangue , Glicoforinas/imunologia , Hemólise , Hipóxia Encefálica/imunologia , Hipóxia Encefálica/patologia , Hipóxia Encefálica/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Camundongos Transgênicos , Circulação Renal , Índice de Gravidade de Doença , Baço/metabolismo , Baço/patologia , Regulação para Cima
16.
Endocr Regul ; 52(3): 152-158, 2018 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-31517605

RESUMO

OBJECTIVE: According to our previous studies, ghrelin protects blood brain barrier (BBB) integrity and it attenuates hypoxia-induced brain edema in the hypoxic conditions. However, the underlying mechanisms remain poorly understood. Several studies suggest a role for matrix metal-loproteinase-9 (MMP9) in the BBB disruption and cerebral edema formation. The present study was conducted to determine the effect of ghrelin on MMP9 protein expression in the model of acute and chronic systemic hypoxia. METHODS: Adult male Wistar rats were divided into acute or chronic controls, acute or chronic hypoxia and ghrelin-treated acute or chronic hypoxia groups. The hypoxic groups were kept in the hypoxic chamber (10-11% O2) for two (acute) or ten days (chronic). Effect of ghrelin on MMP9 protein expression was assessed using immunoblotting. RESULTS: Our results showed that acute and chronic systemic hypoxia increased the MMP9 protein expression in the brain (p<0.001). Treatment with ghrelin significantly attenuated this expression in the cerebral hypoxia (p<0.05). CONCLUSION: Our results demonstrate that the neuroprotective effects of ghrelin may be mediated, in part, by decreasing in MMP9 production in the hypoxic brain.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Grelina/farmacologia , Hipóxia/genética , Metaloproteinase 9 da Matriz/genética , Animais , Encéfalo/patologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Grelina/administração & dosagem , Hipóxia/metabolismo , Hipóxia/patologia , Hipóxia Encefálica/genética , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Neuroproteção/efeitos dos fármacos , Neuroproteção/genética , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar
17.
J Neurochem ; 137(3): 446-59, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26788931

RESUMO

We previously reported that conventional protein kinase C (cPKC)ß participated in hypoxic preconditioning-induced neuroprotection against cerebral ischemic injury, and collapsin response-mediated protein 2 (CRMP2) was identified as a cPKCß interacting protein. In this study, we explored the regulation of CRMP2 phosphorylation and proteolysis by cPKCß, and their role in ischemic injury of oxygen-glucose deprivation (OGD)-treated cortical neurons and brains of mice with middle cerebral artery occlusion-induced ischemic stroke. The results demonstrated that cPKCß-mediated CRMP2 phosphorylation via the cPKCß-selective activator 12-deoxyphorbol 13-phenylacetate 20-acetate (DOPPA) and inhibition of calpain-mediated CRMP2 proteolysis by calpeptin and a fusing peptide containing TAT peptide and the calpain cleavage site of CRMP2 (TAT-CRMP2) protected neurons against OGD-induced cell death through inhibiting CRMP2 proteolysis in cultured cortical neurons. The OGD-induced nuclear translocation of the CRMP2 breakdown product was inhibited by DOPPA, calpeptin, and TAT-CRMP2 in cortical neurons. In addition, both cPKCß activation and CRMP2 proteolysis inhibition by hypoxic preconditioning and intracerebroventricular injections of DOPPA, calpeptin, and TAT-CRMP2 improved the neurological deficit in addition to reducing the infarct volume and proportions of cells with pyknotic nuclei in the peri-infact region of mice with ischemic stroke. These results suggested that cPKCß modulates CRMP2 phosphorylation and proteolysis, and cPKCß activation alleviates ischemic injury in the cultured cortical neurons and brains of mice with ischemic stroke through inhibiting CRMP2 proteolysis by phosphorylation. Focal cerebral ischemia induces a large flux of Ca(2+) to activate calpain which cleaves collapsin response mediator (CRMP) 2 into breakdown product (BDP). Inhibition of CRMP2 cleavage by calpeptin and TAT-CRMP2 alleviates ischemic injury. Conventional protein kinase C (cPKC)ß-mediated phosphorylation could inhibit CRMP2 proteolysis and alleviate ischemic injury in cultured cortical neurons and ischemic stroke-induced mice.


Assuntos
Isquemia Encefálica/patologia , Córtex Cerebral/patologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neurônios/patologia , Proteína Quinase C beta/metabolismo , Acidente Vascular Cerebral/patologia , Animais , Calpaína/antagonistas & inibidores , Células Cultivadas , Glucose/deficiência , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Injeções Intraventriculares , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Precondicionamento Isquêmico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Tecido Nervoso/metabolismo , Ésteres de Forbol/administração & dosagem , Ésteres de Forbol/farmacologia , Fosforilação
18.
Neurobiol Dis ; 90: 68-74, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26777666

RESUMO

We have established that mitochondrial dynamics, under metabolic control, play crucial roles in the regulation of systemic metabolism by hypothalamic circuits. The role of mitochondrial dynamics in neurons in higher brain regions is, however, ill-defined. Here we show that early postnatal exposure of animals to temporal hypoxia followed by normoxia, a major metabolic challenge on brain cells, resulted in adaptive responses of neuronal mitochondria. The number and oxygen consumption of mitochondria in cortical and hippocampal neurons were altered, while mitochondria-endoplasmic reticulum (ER) interactions were preserved. These changes coincided with increased synaptic input of neurons in the cortex and hippocampus. We identified that the changing oxygen tension triggered mitochondrial uncoupling protein 2 (UCP2) expression and showed that UCP2 is crucial for these adaptive mitochondrial responses. In UCP2 KO mice, changing oxygen tension did not induce changes in mitochondrial parameters and function but decreased mitochondria-ER contacts and resulted in loss of synapses both in the cortex and hippocampus. These observations establish that mitochondrial location controlled by UCP2 is relevant for adaptive responses of neurons in cortical and hippocampal neurons and are relevant to perinatal hypoxia-triggered circuit adaptations.


Assuntos
Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Hipóxia Encefálica/metabolismo , Mitocôndrias/metabolismo , Sinapses/metabolismo , Proteína Desacopladora 2/metabolismo , Animais , Contagem de Células , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/patologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/patologia , Hipóxia Encefálica/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Mitocôndrias/patologia , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Neurônios/patologia , Tamanho do Órgão , Sinapses/patologia , Proteína Desacopladora 2/genética
19.
Adv Exp Med Biol ; 903: 201-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27343098

RESUMO

Astrocytes provide the structural and functional interface between the cerebral circulation and neuronal networks. They enwrap all intracerebral arterioles and capillaries, control the flux of nutrients as well as the ionic and metabolic environment of the neuropil. Astrocytes have the ability to adjust cerebral blood flow to maintain constant PO2 and PCO2 of the brain parenchyma. Release of ATP in the brainstem, presumably by local astrocytes, helps to maintain breathing and counteract hypoxia-induced depression of the respiratory network. Astrocytes also appear to be involved in mediating hypoxia-evoked changes in blood-brain barrier permeability, brain inflammation, and neuroprotection against ischaemic injury. Thus, astrocytes appear to play a fundamental role in supporting neuronal function not only in normal conditions but also in pathophysiological states when supply of oxygen to the brain is compromised.


Assuntos
Astrócitos/patologia , Hipóxia Encefálica/patologia , Animais , Barreira Hematoencefálica/metabolismo , Humanos , Inflamação/patologia , Neuroproteção , Acoplamento Neurovascular
20.
Genet Mol Res ; 15(2)2016 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-27323011

RESUMO

Brain damage caused by perinatal asphyxia is dangerous for neonatal infants, but the mechanism by which it occurs remains elusive. In this study, microRNA-152 (miR-152) expression was induced by low oxygen levels in rat models of hypoxia brain damage, as well as in human brain microvascular endothelial cells (HBMECs) cultured in vitro. Analysis of the sequence of miR-152 revealed that the phosphatase and tensin homolog gene (PTEN) is probably the target of miR-152 both in humans and rats. When HBMECs were transfected with miR-152 mimics, PTEN expression was inhibited at both the mRNA and protein levels. Moreover, transfection with the miR-152 mimic also inhibited apoptosis induced by hypoxia. Furthermore, expression of the pro-apoptotic gene Bax was downregulated while the anti-apoptotic gene Bcl2 was upregulated after miR-152 mimic transfection. Taken together, these results indicate that miR-152 induced by hypoxia suppresses cell apoptosis and acts as a protective factor during hypoxia by repressing PTEN.


Assuntos
Células Endoteliais/enzimologia , Hipóxia Encefálica/metabolismo , MicroRNAs/biossíntese , Oxigênio/metabolismo , PTEN Fosfo-Hidrolase/genética , Animais , Apoptose/genética , Encéfalo/irrigação sanguínea , Hipóxia Celular/fisiologia , Células Endoteliais/metabolismo , Células HEK293 , Humanos , Hipóxia Encefálica/patologia , Masculino , Microvasos/enzimologia , Microvasos/metabolismo , Modelos Animais , PTEN Fosfo-Hidrolase/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA