Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Crit Care ; 27(1): 491, 2023 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-38098060

RESUMO

BACKGROUND: Brain injury is a leading cause of morbidity and mortality in patients resuscitated from cardiac arrest. Mitochondrial dysfunction contributes to brain injury following cardiac arrest; therefore, therapies that limit mitochondrial dysfunction have the potential to improve neurological outcomes. Generation of reactive oxygen species (ROS) during ischemia-reperfusion injury in the brain is a critical component of mitochondrial injury and is dependent on hyperactivation of mitochondria following resuscitation. Our previous studies have provided evidence that modulating mitochondrial function with specific near-infrared light (NIR) wavelengths can reduce post-ischemic mitochondrial hyperactivity, thereby reducing brain injury during reperfusion in multiple small animal models. METHODS: Isolated porcine brain cytochrome c oxidase (COX) was used to investigate the mechanism of NIR-induced mitochondrial modulation. Cultured primary neurons from mice expressing mitoQC were utilized to explore the mitochondrial mechanisms related to protection with NIR following ischemia-reperfusion. Anesthetized pigs were used to optimize the delivery of NIR to the brain by measuring the penetration depth of NIR to deep brain structures and tissue heating. Finally, a model of out-of-hospital cardiac arrest with CPR in adult pigs was used to evaluate the translational potential of NIR as a noninvasive therapeutic approach to protect the brain after resuscitation. RESULTS: Molecular evaluation of enzyme activity during NIR irradiation demonstrated COX function was reduced in an intensity-dependent manner with a threshold of enzyme inhibition leading to a moderate reduction in activity without complete inhibition. Mechanistic interrogation in neurons demonstrated that mitochondrial swelling and upregulation of mitophagy were reduced with NIR treatment. NIR therapy in large animals is feasible, as NIR penetrates deep into the brain without substantial tissue heating. In a translational porcine model of CA/CPR, transcranial NIR treatment for two hours at the onset of return of spontaneous circulation (ROSC) demonstrated significantly improved neurological deficit scores and reduced histologic evidence of brain injury after resuscitation from cardiac arrest. CONCLUSIONS: NIR modulates mitochondrial function which improves mitochondrial dynamics and quality control following ischemia/reperfusion. Noninvasive modulation of mitochondria, achieved by transcranial treatment of the brain with NIR, mitigates post-cardiac arrest brain injury and improves neurologic functional outcomes.


Assuntos
Lesões Encefálicas , Reanimação Cardiopulmonar , Doenças Mitocondriais , Parada Cardíaca Extra-Hospitalar , Humanos , Camundongos , Animais , Suínos , Mitocôndrias , Isquemia , Modelos Animais de Doenças
2.
Pediatr Neurol ; 145: 57-66, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37279616

RESUMO

BACKGROUND: A relative shortage of pediatric neurologists in proportion to estimated neurological disorders often results in general pediatricians evaluating and treating children with complex neurological conditions. Dedicated rotations in pediatric neurology are not mandated during medical school or pediatric residency. We evaluated the perceptions of a large cohort of pediatric residents and program directors (PDs) regarding child neurology training. METHODS: Using an online tool, surveys were sent to pediatric residents and pediatric and pediatric neurology PDs. RESULTS: Response rates were 41% from pediatric residency programs, yielding 538 resident responses; 31% from pediatric PDs; and 62% from pediatric neurology PDs. Only 27% of the surveyed residents reported completing a neurology rotation during residency, 89% of whom expressed a subjective improvement in confidence with neurological assessments. Factors affecting comfort with eliciting a neurological history included exposure to a neurology rotation during residency, year of training, duration of neurology rotation in medical school, and inpatient exposure to neurological patients, whereas those associated with examination additionally included program size and postresidency plans. Overall, 80% of surveyed residents, 78% of pediatric PDs, and 96% of pediatric neurology PDs acknowledged the potential value of a mandatory pediatric neurology rotation during residency. CONCLUSION: We suggest that a mandatory pediatric neurology rotation will boost the confidence of current and future pediatric trainees in assessing common neurological conditions of childhood.


Assuntos
Internato e Residência , Neurologia , Humanos , Criança , Estados Unidos , Educação de Pós-Graduação em Medicina , Neurologia/educação , Neurologistas , Currículo , Inquéritos e Questionários
3.
Cells ; 11(19)2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36231044

RESUMO

Disruption of mitochondrial structure/function is well-recognized to be a determinant of cell death in cardiomyocytes subjected to lethal episodes of ischemia-reperfusion (IR). However, the precise mitochondrial event(s) that precipitate lethal IR injury remain incompletely resolved. Using the in vitro HL-1 cardiomyocyte model, our aims were to establish whether: (1) proteolytic processing of optic atrophy protein-1 (OPA1), the inner mitochondrial membrane protein responsible for maintaining cristae junction integrity, plays a causal, mechanistic role in determining cardiomyocyte fate in cells subjected to lethal IR injury; and (2) preservation of OPA1 may contribute to the well-documented cardioprotection achieved with ischemic preconditioning (IPC) and remote ischemic conditioning. We report that HL-1 cells subjected to 2.5 h of simulated ischemia displayed increased activity of OMA1 (the metalloprotease responsible for proteolytic processing of OPA1) during the initial 45 min following reoxygenation. This was accompanied by processing of mitochondrial OPA1 (i.e., cleavage to yield short-OPA1 peptides) and release of short-OPA1 into the cytosol. However, siRNA-mediated knockdown of OPA1 content did not exacerbate lethal IR injury, and did not attenuate the cardioprotection seen with IPC and a remote preconditioning stimulus, achieved by transfer of 'reperfusate' medium (TRM-IPC) in this cell culture model. Taken together, our results do not support the concept that maintenance of OPA1 integrity plays a mechanistic role in determining cell fate in the HL-1 cardiomyocyte model of lethal IR injury, or that preservation of OPA1 underlies the cardioprotection seen with ischemic conditioning.


Assuntos
Atrofia Óptica , Traumatismo por Reperfusão , Morte Celular , GTP Fosfo-Hidrolases/metabolismo , Humanos , Isquemia/metabolismo , Metaloproteases/metabolismo , Miócitos Cardíacos/metabolismo , Atrofia Óptica/metabolismo , RNA Interferente Pequeno/metabolismo , Traumatismo por Reperfusão/metabolismo
4.
J Clin Med ; 11(9)2022 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-35566399

RESUMO

Parathyroid hormone-related peptide (PTHrP) is well-known to play a role in bone formation, and abaloparatide, an analog of PTHrP(1-34), is approved for the treatment of osteoporosis in post-menopausal women. PTHrP has also been reported to have cardiovascular effects, with recent data demonstrating that exogenously administered PTHrP can limit the death of isolated cardiomyocytes subjected to oxidative stress via upregulation of classic 'survival kinase' signaling. Our aim in the current study was to extend this concept and, employing both in vitro and in vivo models, establish whether PTHrP(1-36) and abaloparatide are cardioprotective in the setting of lethal myocardial ischemia-reperfusion injury. We report that preischemic administration of PTHrP(1-36) and abaloparatide attenuated cell death in HL-1 cardiomyocytes subjected to simulated ischemia-reperfusion, an effect that was accompanied by the augmented expression of phospho-ERK and improved preservation of phospho-Akt, and blocked by co-administration of the MEK-ERK inhibitor PD98059. Moreover, using the translationally relevant swine model of acute coronary artery occlusion-reperfusion, we make the novel observation that myocardial infarct size was significantly reduced in pigs pretreated with PTHrP(1-36) when compared with placebo-controls (13.1 ± 3.3% versus 42.0 ± 6.6% of the area of at-risk myocardium, respectively; p < 0.01). Taken together, these data provide the first evidence in support of the concept that pretreatment with PTHrP(1-36) and abaloparatide renders cardiomyocytes resistant to lethal myocardial ischemia-reperfusion injury.

5.
Cell Death Dis ; 12(5): 475, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980811

RESUMO

Mitochondrial dynamics and mitophagy are constitutive and complex systems that ensure a healthy mitochondrial network through the segregation and subsequent degradation of damaged mitochondria. Disruption of these systems can lead to mitochondrial dysfunction and has been established as a central mechanism of ischemia/reperfusion (I/R) injury. Emerging evidence suggests that mitochondrial dynamics and mitophagy are integrated systems; however, the role of this relationship in the context of I/R injury remains unclear. To investigate this concept, we utilized primary cortical neurons isolated from the novel dual-reporter mitochondrial quality control knockin mice (C57BL/6-Gt(ROSA)26Sortm1(CAG-mCherry/GFP)Ganl/J) with conditional knockout (KO) of Drp1 to investigate changes in mitochondrial dynamics and mitophagic flux during in vitro I/R injury. Mitochondrial dynamics was quantitatively measured in an unbiased manner using a machine learning mitochondrial morphology classification system, which consisted of four different classifications: network, unbranched, swollen, and punctate. Evaluation of mitochondrial morphology and mitophagic flux in primary neurons exposed to oxygen-glucose deprivation (OGD) and reoxygenation (OGD/R) revealed extensive mitochondrial fragmentation and swelling, together with a significant upregulation in mitophagic flux. Furthermore, the primary morphology of mitochondria undergoing mitophagy was classified as punctate. Colocalization using immunofluorescence as well as western blot analysis revealed that the PINK1/Parkin pathway of mitophagy was activated following OGD/R. Conditional KO of Drp1 prevented mitochondrial fragmentation and swelling following OGD/R but did not alter mitophagic flux. These data provide novel evidence that Drp1 plays a causal role in the progression of I/R injury, but mitophagy does not require Drp1-mediated mitochondrial fission.


Assuntos
Dinaminas/metabolismo , Dinâmica Mitocondrial/genética , Mitofagia/genética , Traumatismo por Reperfusão/genética , Animais , Humanos , Camundongos , Neurônios/metabolismo , Traumatismo por Reperfusão/metabolismo
6.
Sci Rep ; 11(1): 5133, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33664336

RESUMO

The mitochondrial network continually undergoes events of fission and fusion. Under physiologic conditions, the network is in equilibrium and is characterized by the presence of both elongated and punctate mitochondria. However, this balanced, homeostatic mitochondrial profile can change morphologic distribution in response to various stressors. Therefore, it is imperative to develop a method that robustly measures mitochondrial morphology with high accuracy. Here, we developed a semi-automated image analysis pipeline for the quantitation of mitochondrial morphology for both in vitro and in vivo applications. The image analysis pipeline was generated and validated utilizing images of primary cortical neurons from transgenic mice, allowing genetic ablation of key components of mitochondrial dynamics. This analysis pipeline was further extended to evaluate mitochondrial morphology in vivo through immunolabeling of brain sections as well as serial block-face scanning electron microscopy. These data demonstrate a highly specific and sensitive method that accurately classifies distinct physiological and pathological mitochondrial morphologies. Furthermore, this workflow employs the use of readily available, free open-source software designed for high throughput image processing, segmentation, and analysis that is customizable to various biological models.


Assuntos
Encéfalo/diagnóstico por imagem , Aprendizado de Máquina , Mitocôndrias/ultraestrutura , Neurônios/ultraestrutura , Animais , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Humanos , Processamento de Imagem Assistida por Computador , Camundongos , Microscopia Eletrônica de Varredura , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Rede Nervosa/diagnóstico por imagem , Neurônios/metabolismo
7.
Cells ; 9(1)2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31952189

RESUMO

The current standard of care for acute myocardial infarction or 'heart attack' is timely restoration of blood flow to the ischemic region of the heart. While reperfusion is essential for the salvage of ischemic myocardium, re-introduction of blood flow paradoxically kills (rather than rescues) a population of previously ischemic cardiomyocytes-a phenomenon referred to as 'lethal myocardial ischemia-reperfusion (IR) injury'. There is long-standing and exhaustive evidence that mitochondria are at the nexus of lethal IR injury. However, during the past decade, the paradigm of mitochondria as mediators of IR-induced cardiomyocyte death has been expanded to include the highly orchestrated process of mitochondrial quality control. Our aims in this review are to: (1) briefly summarize the current understanding of the pathogenesis of IR injury, and (2) incorporating landmark data from a broad spectrum of models (including immortalized cells, primary cardiomyocytes and intact hearts), provide a critical discussion of the emerging concept that mitochondrial dynamics and mitophagy (the components of mitochondrial quality control) may contribute to the pathogenesis of cardiomyocyte death in the setting of ischemia-reperfusion.


Assuntos
Mitocôndrias Cardíacas/metabolismo , Modelos Cardiovasculares , Traumatismo por Reperfusão/patologia , Animais , Encéfalo/patologia , Humanos , Dinâmica Mitocondrial , Miócitos Cardíacos/patologia
8.
J Cereb Blood Flow Metab ; 40(4): 833-844, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31112450

RESUMO

Ischemic stroke is a debilitating disease that causes significant brain injury. While restoration of blood flow is critical to salvage the ischemic brain, reperfusion can exacerbate damage by inducing generation of reactive oxygen species (ROS). Recent studies by our group found that non-invasive mitochondrial modulation with near-infrared (NIR) light limits ROS generation following global brain ischemia. NIR interacts with cytochrome c oxidase (COX) to transiently reduce COX activity, attenuate mitochondrial membrane potential hyperpolarization, and thus reduce ROS production. We evaluated a specific combination of COX-inhibitory NIR (750 nm and 950 nm) in a rat stroke model with longitudinal analysis of brain injury using magnetic resonance imaging. Treatment with NIR for 2 h resulted in a 21% reduction in brain injury at 24 h of reperfusion measured by diffusion-weighted imaging (DWI) and a 25% reduction in infarct volume measured by T2-weighted imaging (T2WI) at 7 and 14 days of reperfusion, respectively. Additionally, extended treatment reduced brain injury in the acute phase of brain injury, and 7 and 14 days of reperfusion, demonstrating a >50% reduction in infarction. Our data suggest that mitochondrial modulation with NIR attenuates ischemia-reperfusion injury and evokes a sustained reduction in infarct volume following ischemic stroke.


Assuntos
Encéfalo/metabolismo , Raios Infravermelhos/uso terapêutico , AVC Isquêmico/terapia , Traumatismo por Reperfusão/prevenção & controle , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos da radiação , Circulação Cerebrovascular/efeitos da radiação , Imagem de Difusão por Ressonância Magnética , Complexo IV da Cadeia de Transporte de Elétrons/antagonistas & inibidores , AVC Isquêmico/complicações , AVC Isquêmico/diagnóstico por imagem , AVC Isquêmico/metabolismo , Potencial da Membrana Mitocondrial/efeitos da radiação , Ratos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/diagnóstico por imagem
10.
Exp Physiol ; 104(3): 278-294, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30597638

RESUMO

NEW FINDINGS: What is the topic of this review? Remote ischaemic preconditioning (RIPC) and hypoxic preconditioning as novel therapeutic approaches for cardiac and neuroprotection. What advances does it highlight? There is improved understanding of mechanisms and signalling pathways associated with ischaemic and hypoxic preconditioning, and potential pitfalls with application of these therapies to clinical trials have been identified. Novel adaptations of preconditioning paradigms have also been developed, including intermittent hypoxia training, RIPC training and RIPC-exercise, extending their utility to chronic settings. ABSTRACT: Myocardial infarction and stroke remain leading causes of death worldwide, despite extensive resources directed towards developing effective treatments. In this Symposium Report we highlight the potential applications of intermittent ischaemic and hypoxic conditioning protocols to combat the deleterious consequences of heart and brain ischaemia. Insights into mechanisms underlying the protective effects of intermittent hypoxia training are discussed, including the activation of hypoxia-inducible factor-1 and Nrf2 transcription factors, synthesis of antioxidant and ATP-generating enzymes, and a shift in microglia from pro- to anti-inflammatory phenotypes. Although there is little argument regarding the efficacy of remote ischaemic preconditioning (RIPC) in pre-clinical models, this strategy has not consistently translated into the clinical arena. This lack of translation may be related to the patient populations targeted thus far, and the anaesthetic regimen used in two of the major RIPC clinical trials. Additionally, we do not fully understand the mechanism through which RIPC protects the vital organs, and co-morbidities (e.g. hypercholesterolemia, diabetes) may interfere with its efficacy. Finally, novel adaptations have been made to extend RIPC to more chronic settings. One adaptation is RIPC-exercise (RIPC-X), an innovative paradigm that applies cyclical RIPC to blood flow restriction exercise (BFRE). Recent findings suggest that this novel exercise modality attenuates the exaggerated haemodynamic responses that may limit the use of conventional BFRE in some clinical settings. Collectively, intermittent ischaemic and hypoxic conditioning paradigms remain an exciting frontier for the protection against ischaemic injuries.


Assuntos
Encéfalo/fisiopatologia , Coração/fisiopatologia , Hipóxia/fisiopatologia , Infarto do Miocárdio/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Exercício Físico/fisiologia , Hemodinâmica/fisiologia , Humanos , Precondicionamento Isquêmico/métodos
11.
Sci Rep ; 8(1): 17580, 2018 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-30498241

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

12.
Sci Rep ; 8(1): 14529, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30266987

RESUMO

Dual antiplatelet therapy with aspirin and an adenosine diphosphate (ADP) P2Y12 receptor antagonist reduces ischemic events in patients with acute coronary syndrome. Previous evidence from our group, obtained in a preclinical model of recurrent platelet-mediated thrombosis, demonstrated that GLS-409, a diadenosine tetraphosphate derivative that inhibits both P2Y1 and P2Y12 ADP receptors, may be a novel and promising antiplatelet drug candidate. However, the salutary antiplatelet effects of GLS-409 were accompanied by a trend toward an unfavorable increase in bleeding. The goals of this study were to: 1) provide proof-of-concept that the efficacy of GLS-409 may be maintained at lower dose(s), not accompanied by an increased propensity to bleeding; and 2) establish the extent and kinetics of the reversibility of human platelet inhibition by the agent. Lower doses of GLS-409 were identified that inhibited in vivo recurrent coronary thrombosis with no increase in bleeding time. Human platelet inhibition by GLS-409 was reversible, with rapid recovery of platelet reactivity to ADP, as measured by platelet surface activated GPIIb-IIIa and platelet surface P-selectin. These data support the concept that GLS-409 warrants further, larger-scale investigation as a novel, potential therapy in acute coronary syndromes.


Assuntos
Trombose Coronária/veterinária , Fosfatos de Dinucleosídeos/uso terapêutico , Doenças do Cão/tratamento farmacológico , Ativação Plaquetária/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Animais , Trombose Coronária/tratamento farmacológico , Trombose Coronária/metabolismo , Doenças do Cão/metabolismo , Cães , Feminino , Humanos , Inibidores da Agregação Plaquetária/uso terapêutico , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2Y12/metabolismo
14.
Basic Res Cardiol ; 113(3): 16, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29524006

RESUMO

Remote ischemic preconditioning (RIPC), the phenomenon whereby brief ischemic episodes in distant tissues or organs render the heart resistant to infarction, has been exhaustively demonstrated in preclinical models. Moreover, emerging evidence suggests that exosomes play a requisite role in conveying the cardioprotective signal from remote tissue to the myocardium. However, in cohorts displaying clinically common comorbidities-in particular, type-2 diabetes-the infarct-sparing effect of RIPC may be confounded for as-yet unknown reasons. To investigate this issue, we used an integrated in vivo and in vitro approach to establish whether: (1) the efficacy of RIPC is maintained in the Zucker fatty rat model of type-2 diabetes, (2) the humoral transfer of cardioprotective triggers initiated by RIPC are transported via exosomes, and (3) diabetes is associated with alterations in exosome-mediated communication. We report that a standard RIPC stimulus (four 5-min episodes of hindlimb ischemia) reduced infarct size in normoglycemic Zucker lean rats, but failed to confer protection in diabetic Zucker fatty animals. Moreover, we provide novel evidence, via transfer of serum and serum fractions obtained following RIPC and applied to HL-1 cardiomyocytes subjected to hypoxia-reoxygenation, that diabetes was accompanied by impaired humoral communication of cardioprotective signals. Specifically, our data revealed that serum and exosome-rich serum fractions collected from normoglycemic rats attenuated hypoxia-reoxygenation-induced HL-1 cell death, while, in contrast, exosome-rich samples from Zucker fatty rats did not evoke protection in the HL-1 cell model. Finally, and unexpectedly, we found that exosome-depleted serum from Zucker fatty rats was cytotoxic and exacerbated hypoxia-reoxygenation-induced cardiomyocyte death.


Assuntos
Diabetes Mellitus Tipo 2/terapia , Exossomos/metabolismo , Membro Posterior/irrigação sanguínea , Precondicionamento Isquêmico/métodos , Infarto do Miocárdio/prevenção & controle , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Animais , Morte Celular , Linhagem Celular , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Exossomos/patologia , Masculino , Infarto do Miocárdio/sangue , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Ratos Zucker , Fluxo Sanguíneo Regional
15.
Sci Rep ; 8(1): 3481, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29472564

RESUMO

The interaction of light with biological tissue has been successfully utilized for multiple therapeutic purposes. Previous studies have suggested that near infrared light (NIR) enhances the activity of mitochondria by increasing cytochrome c oxidase (COX) activity, which we confirmed for 810 nm NIR. In contrast, scanning the NIR spectrum between 700 nm and 1000 nm revealed two NIR wavelengths (750 nm and 950 nm) that reduced the activity of isolated COX. COX-inhibitory wavelengths reduced mitochondrial respiration, reduced the mitochondrial membrane potential (ΔΨm), attenuated mitochondrial superoxide production, and attenuated neuronal death following oxygen glucose deprivation, whereas NIR that activates COX provided no benefit. We evaluated COX-inhibitory NIR as a potential therapy for cerebral reperfusion injury using a rat model of global brain ischemia. Untreated animals demonstrated an 86% loss of neurons in the CA1 hippocampus post-reperfusion whereas inhibitory NIR groups were robustly protected, with neuronal loss ranging from 11% to 35%. Moreover, neurologic function, assessed by radial arm maze performance, was preserved at control levels in rats treated with a combination of both COX-inhibitory NIR wavelengths. Taken together, our data suggest that COX-inhibitory NIR may be a viable non-pharmacologic and noninvasive therapy for the treatment of cerebral reperfusion injury.


Assuntos
Lesões Encefálicas/radioterapia , Complexo IV da Cadeia de Transporte de Elétrons/genética , Raios Infravermelhos/uso terapêutico , Traumatismo por Reperfusão/radioterapia , Animais , Encéfalo/patologia , Encéfalo/efeitos da radiação , Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Complexo IV da Cadeia de Transporte de Elétrons/efeitos da radiação , Glucose/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/efeitos da radiação , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias/genética , Mitocôndrias/efeitos da radiação , Neurônios/metabolismo , Neurônios/efeitos da radiação , Oxirredução/efeitos da radiação , Ratos , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
16.
Am J Physiol Heart Circ Physiol ; 314(4): H812-H838, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351451

RESUMO

Myocardial infarction is a prevalent major cardiovascular event that arises from myocardial ischemia with or without reperfusion, and basic and translational research is needed to better understand its underlying mechanisms and consequences for cardiac structure and function. Ischemia underlies a broad range of clinical scenarios ranging from angina to hibernation to permanent occlusion, and while reperfusion is mandatory for salvage from ischemic injury, reperfusion also inflicts injury on its own. In this consensus statement, we present recommendations for animal models of myocardial ischemia and infarction. With increasing awareness of the need for rigor and reproducibility in designing and performing scientific research to ensure validation of results, the goal of this review is to provide best practice information regarding myocardial ischemia-reperfusion and infarction models. Listen to this article's corresponding podcast at ajpheart.podbean.com/e/guidelines-for-experimental-models-of-myocardial-ischemia-and-infarction/.


Assuntos
Pesquisa Biomédica/normas , Cardiologia/normas , Infarto do Miocárdio , Isquemia Miocárdica , Publicações Periódicas como Assunto/normas , Fisiologia/normas , Animais , Células Cultivadas , Consenso , Confiabilidade dos Dados , Modelos Animais de Doenças , Preparação de Coração Isolado/normas , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Controle de Qualidade
17.
Mol Neurobiol ; 55(3): 2547-2564, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28401475

RESUMO

Mitochondria are key regulators of cell fate during disease. They control cell survival via the production of ATP that fuels cellular processes and, conversely, cell death via the induction of apoptosis through release of pro-apoptotic factors such as cytochrome C. Therefore, it is essential to have stringent quality control mechanisms to ensure a healthy mitochondrial network. Quality control mechanisms are largely regulated by mitochondrial dynamics and mitophagy. The processes of mitochondrial fission (division) and fusion allow for damaged mitochondria to be segregated and facilitate the equilibration of mitochondrial components such as DNA, proteins, and metabolites. The process of mitophagy are responsible for the degradation and recycling of damaged mitochondria. These mitochondrial quality control mechanisms have been well studied in chronic and acute pathologies such as Parkinson's disease, Alzheimer's disease, stroke, and acute myocardial infarction, but less is known about how these two processes interact and contribute to specific pathophysiologic states. To date, evidence for the role of mitochondrial quality control in acute and chronic disease is divergent and suggests that mitochondrial quality control processes can serve both survival and death functions depending on the disease state. This review aims to provide a synopsis of the molecular mechanisms involved in mitochondrial quality control, to summarize our current understanding of the complex role that mitochondrial quality control plays in the progression of acute vs chronic diseases and, finally, to speculate on the possibility that targeted manipulation of mitochondrial quality control mechanisms may be exploited for the rationale design of novel therapeutic interventions.


Assuntos
Mitocôndrias/metabolismo , Doenças Neurodegenerativas/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Humanos , Mitocôndrias/genética , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Controle de Qualidade , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
18.
J Cardiovasc Pharmacol Ther ; 22(5): 391-396, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28793820

RESUMO

Data obtained in both preclinical models and humans have revealed that the favorable cardiac consequences of ischemic conditioning extend beyond a direct effect on the cardiomyocyte. In the current review, we summarize our as-yet limited understanding of the complex relationships between ischemic conditioning, platelet activation-aggregation, and cardioprotection.


Assuntos
Plaquetas/fisiologia , Precondicionamento Isquêmico Miocárdico , Trombose/prevenção & controle , Humanos , Ativação Plaquetária , Antagonistas do Receptor Purinérgico P2Y/farmacologia
19.
BMJ Open ; 7(8): e016479, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28814584

RESUMO

OBJECTIVES: To examine the relationship between perceived and biological stress and near misses among Emergency Medicine residents. DESIGN: Self-rated stress and stress biomarkers were assessed in residents in Emergency Medicine before and after a day shift. The supervising physicians and residents reported numbers of near misses. SETTING: The study took place in the Emergency Department of a large trauma 1 centre, located in Detroit, USA. PARTICIPANTS: Residents in Emergency Medicine volunteered to participate. The sample consisted of 32 residents, with complete data on 28 subjects. Residents' supervising physicians assessed the clinical performance of each resident. PRIMARY AND SECONDARY OUTCOME MEASURES: Participants' preshift and postshift stress, biological stress (salivary cortisol, plasma interleukin-6, tumour necrosis factor-alpha (TNF-α) and high-sensitivity C-reactive protein), residents' and supervisors' reports of near misses, number of critically ill and patients with trauma seen during the shift. RESULTS: Residents' self-reported stress increased from an average preshift level of 2.79 of 10 (SD 1.81) to a postshift level of 5.82 (2.13) (p<0.001). Residents cared for an average of 2.32 (1.52) critically ill patients and 0.68 (1.06) patients with trauma. Residents reported a total of 7 near misses, compared with 11 reported by the supervising physicians. After controlling for baseline work-related exhaustion, residents that cared for more patients with trauma and had higher levels of TNF-α reported a higher frequency of near misses (R2=0.72; p=0.001). Residents' preshift ratings of how stressful they expected the shift to be were related to the supervising physicians' ratings of residents' near misses during the shift. CONCLUSION: Residents' own ratings of near misses were associated with residents' TNF-α, a biomarker of systemic inflammation and the number of patients with trauma seen during the shift. In contrast, supervisor reports on residents' near misses were related only to the residents' preshift expectations of how stressful the shift would be.


Assuntos
Competência Clínica/normas , Tomada de Decisões/fisiologia , Medicina de Emergência , Internato e Residência , Near Miss/estatística & dados numéricos , Estresse Fisiológico/fisiologia , Tolerância ao Trabalho Programado/fisiologia , Adulto , Proteína C-Reativa/análise , Estudos Transversais , Avaliação Educacional , Feminino , Humanos , Hidrocortisona/análise , Interleucina-6/sangue , Masculino , Autorrelato , Fator de Necrose Tumoral alfa/sangue , Estados Unidos , Tolerância ao Trabalho Programado/psicologia , Carga de Trabalho
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...