Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cardiooncology ; 9(1): 36, 2023 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-37803479

RESUMO

OBJECTIVE: To determine the impact of acute SARS-CoV-2 infection on patient with concomitant active cancer and CVD. METHODS: The researchers extracted and analyzed data from the National COVID Cohort Collaborative (N3C) database between January 1, 2020, and July 22, 2022. They included only patients with acute SARS-CoV-2 infection, defined as a positive test by PCR 21 days before and 5 days after the day of index hospitalization. Active cancers were defined as last cancer drug administered within 30 days of index admission. The "Cardioonc" group consisted of patients with CVD and active cancers. The cohort was divided into four groups: (1) CVD (-), (2) CVD ( +), (3) Cardioonc (-), and (4) Cardioonc ( +), where (-) or ( +) denotes acute SARS-CoV-2 infection status. The primary outcome of the study was major adverse cardiovascular events (MACE), including acute stroke, acute heart failure, myocardial infarction, or all-cause mortality. The researchers analyzed the outcomes by different phases of the pandemic and performed competing-risk analysis for other MACE components and death as a competing event. RESULTS: The study analyzed 418,306 patients, of which 74%, 10%, 15.7%, and 0.3% had CVD (-), CVD ( +), Cardioonc (-), and Cardioonc ( +), respectively. The Cardioonc ( +) group had the highest MACE events in all four phases of the pandemic. Compared to CVD (-), the Cardioonc ( +) group had an odds ratio of 1.66 for MACE. However, during the Omicron era, there was a statistically significant increased risk for MACE in the Cardioonc ( +) group compared to CVD (-). Competing risk analysis showed that all-cause mortality was significantly higher in the Cardioonc ( +) group and limited other MACE events from occurring. When the researchers identified specific cancer types, patients with colon cancer had higher MACE. CONCLUSION: In conclusion, the study found that patients with both CVD and active cancer suffered relatively worse outcomes when they had acute SARS-CoV-2 infection during early and alpha surges in the United States. These findings highlight the need for improved management strategies and further research to better understand the impact of the virus on vulnerable populations during the COVID-19 pandemic.

2.
J Cell Physiol ; 2022 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-36036549

RESUMO

Alzheimer's disease (AD) is the leading cause of mortality, disability, and long-term care burden in the United States, with women comprising the majority of AD diagnoses. While AD-related dementia is associated with tau and amyloid beta accumulation, concurrent derangements in cerebral blood flow have been observed alongside these proteinopathies in humans and rodent models. The homeostatic production of nitric oxide synthases (NOS) becomes uncoupled in AD which leads to decreased NO-mediated vasodilation and oxidative stress via the production of peroxynitrite (ONOO-∙) superoxide species. Here, we investigate the role of the novel protein arginine methyltransferase 4 (PRMT4) enzyme function and its downstream product asymmetric dimethyl arginine (ADMA) as it relates to NOS dysregulation and cerebral blood flow in AD. ADMA (type-1 PRMT product) has been shown to bind NOS as a noncanonic ligand causing enzymatic dysfunction. Our results from RT-qPCR and protein analyses suggest that aged (9-12 months) female mice bearing tau- and amyloid beta-producing transgenic mutations (3xTg-AD) express higher levels of PRMT4 in the hippocampus when compared to age- and sex-matched C57BL6/J mice. In addition, we performed studies to quantify the expression and activity of different NOS isoforms. Furthermore, laser speckle contrast imaging analysis was indicative that 3xTg-AD mice have dysfunctional NOS activity, resulting in reduced production of NO metabolites, enhanced production of free-radical ONOO-, and decreased cerebral blood flow. Notably, the aforementioned phenomena can be reversed via pharmacologic PRMT4 inhibition. Together, these findings implicate the potential importance of PRMT4 signaling in the pathogenesis of Alzheimer's-related cerebrovascular derangement.

3.
Neuromolecular Med ; 24(2): 97-112, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34019239

RESUMO

Cardiopulmonary arrest (CA) can greatly impact a patient's life, causing long-term disability and death. Although multi-faceted treatment strategies against CA have improved survival rates, the prognosis of CA remains poor. We previously reported asphyxial cardiac arrest (ACA) can cause excessive activation of the sympathetic nervous system (SNS) in the brain, which contributes to cerebral blood flow (CBF) derangements such as hypoperfusion and, consequently, neurological deficits. Here, we report excessive activation of the SNS can cause enhanced neuropeptide Y levels. In fact, mRNA and protein levels of neuropeptide Y (NPY, a 36-amino acid neuropeptide) in the hippocampus were elevated after ACA-induced SNS activation, resulting in a reduced blood supply to the brain. Post-treatment with peptide YY3-36 (PYY3-36), a pre-synaptic NPY2 receptor agonist, after ACA inhibited NPY release and restored brain circulation. Moreover, PYY3-36 decreased neuroinflammatory cytokines, alleviated mitochondrial dysfunction, and improved neuronal survival and neurological outcomes. Overall, NPY is detrimental during/after ACA, but attenuation of NPY release via PYY3-36 affords neuroprotection. The consequences of PYY3-36 inhibit ACA-induced 1) hypoperfusion, 2) neuroinflammation, 3) mitochondrial dysfunction, 4) neuronal cell death, and 5) neurological deficits. The present study provides novel insights to further our understanding of NPY's role in ischemic brain injury.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , Parada Cardíaca , Animais , Lesões Encefálicas/etiologia , Isquemia Encefálica/complicações , Parada Cardíaca/complicações , Parada Cardíaca/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeo Y/agonistas , Receptores de Neuropeptídeo Y/genética
4.
Am J Physiol Heart Circ Physiol ; 319(5): H1044-H1050, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32946263

RESUMO

Cardiopulmonary arrest (CA) is the leading cause of death and disability in the United States. CA-induced brain injury is influenced by multifactorial processes, including reduced cerebral blood flow (hypoperfusion) and neuroinflammation, which can lead to neuronal cell death and functional deficits. We have identified serum and glucocorticoid-regulated kinase-1 (SGK1) as a new target in brain ischemia previously described in the heart, liver, and kidneys (i.e., diabetes and hypertension). Our data suggest brain SGK1 mRNA and protein expression (i.e., hippocampus), presented with hypoperfusion (low cerebral blood flow) and neuroinflammation, leading to further studies of the potential role of SGK1 in CA-induced brain injury. We used a 6-min asphyxia cardiac arrest (ACA) rat model to induce global cerebral ischemia. Modulation of SGK1 was implemented via GSK650394, a SGK1-specific inhibitor (1.2 µg/kg icv). Accordingly, treatment with GSK650394 attenuated cortical hypoperfusion and neuroinflammation (via Iba1 expression) after ACA, whereas neuronal survival was enhanced in the CA1 region of the hippocampus. Learning/memory deficits were observed 3 days after ACA but ameliorated with GSK650394. In conclusion, SGK1 is a major contributor to ACA-induced brain injury and neurological deficits, while inhibition of SGK1 with GSK650394 provided neuroprotection against CA-induced hypoperfusion, neuroinflammation, neuronal cell death, and learning/memory deficits. Our studies could lead to a novel, therapeutic target for alleviating brain injury following cerebral ischemia.NEW & NOTEWORTHY Upregulation of SGK1 exacerbates brain injury during cerebral ischemia. Inhibition of SGK1 affords neuroprotection against cardiac arrest-induced hypoperfusion, neuroinflammation, neuronal cell death, and neurological deficits.


Assuntos
Lesões Encefálicas/metabolismo , Parada Cardíaca/complicações , Proteínas Imediatamente Precoces/genética , Memória , Proteínas Serina-Treonina Quinases/genética , Animais , Benzoatos/farmacologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/etiologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Circulação Cerebrovascular , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/metabolismo , Masculino , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima
5.
Artigo em Inglês | MEDLINE | ID: mdl-32663656

RESUMO

Cardiac arrest causes neuronal damage and functional impairments that can result in learning/memory dysfunction after ischemia. We previously identified a saturated fatty acid (stearic acid methyl ester, SAME) that was released from the superior cervical ganglion (sympathetic ganglion). The function of stearic acid methyl ester is currently unknown. Here, we show that SAME can inhibit the detrimental effects of global cerebral ischemia (i.e. cardiac arrest). Treatment with SAME in the presence of asphyxial cardiac arrest (ACA) revived learning and working memory deficits. Similarly, SAME-treated hippocampal slices after oxygen-glucose deprivation inhibited neuronal cell death. Moreover, SAME afforded neuroprotection against ACA in the CA1 region of the hippocampus, reduced ionized calcium-binding adapter molecule 1 expression and inflammatory cytokines/chemokines, with restoration in mitochondria respiration. Altogether, we describe a unique and uncharted role of saturated fatty acids in the brain that may have important implications against cerebral ischemia.


Assuntos
Asfixia/tratamento farmacológico , Região CA1 Hipocampal/metabolismo , Parada Cardíaca/tratamento farmacológico , Neuroproteção/efeitos dos fármacos , Ácidos Esteáricos/farmacologia , Animais , Asfixia/metabolismo , Asfixia/fisiopatologia , Região CA1 Hipocampal/fisiopatologia , Modelos Animais de Doenças , Parada Cardíaca/metabolismo , Parada Cardíaca/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley
6.
Artigo em Inglês | MEDLINE | ID: mdl-32447175

RESUMO

Sickle cell disease (SCD) is one of the most common inherited blood disorder among African Americans affecting 70,000-100,000 individuals in the United States. It is characterized by abnormal hemoglobin (HbS) which develops into severe hemolytic anemia and vaso-occlusive crisis. Therefore, patients with SCD suffer from a chronic state of inflammation, which is responsible for multiple organ damage, ischemic attacks, and premature death. Another major hallmark of SCD patients is the abnormally low levels of omega-3 fatty acids, especially docosahexaenoic acid (DHA) in their red blood cell membranes. Treatment with DHA can reduce red blood cell adhesion and enhance cerebral blood flow, thus, our main goal is to investigate the effect of SC411, which is a novel, highly purified DHA ethyl ester formulation with a proprietary delivery platform in SCD. Utilizing a transgenic mouse model of SCD (HbSS-Townes) and recurrent hypoxic challenges (10%O2, 0.5% CO2 and balance N2 for 3 h) to mimic ischemic-like conditions, our data suggest that SC411 can elevate blood DHA and eicosapentaenoic acid (EPA) levels after 8 weeks of treatment. SC411 can also decrease arachidonic acid (AA) and sickling of red blood cells. In addition, SC411-treated SCD mice showed presented with cerebral blood flow, alleviated neuroinflammation, and revived working memory which ultimately enhanced overall survival. In summary, this study suggests that treatment with SC411 improves cellular and functional outcomes in SCD mice. This finding may provide novel therapeutic opportunities in the treatment against ischemic injury elicited by SCD.


Assuntos
Anemia Falciforme/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/química , Ésteres/administração & dosagem , Anemia Falciforme/genética , Anemia Falciforme/psicologia , Animais , Ácido Araquidônico/sangue , Circulação Cerebrovascular , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/sangue , Ésteres/química , Ésteres/farmacologia , Humanos , Masculino , Memória de Curto Prazo/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Análise de Sobrevida , Resultado do Tratamento
7.
Artigo em Inglês | MEDLINE | ID: mdl-30514597

RESUMO

We previously discovered that palmitic acid methyl ester (PAME) is a potent vasodilator first identified and released from the superior cervical ganglion and remain understudied. Thus, we investigated PAME's role in modulating cerebral blood flow (CBF) and neuroprotection after 6 min of cardiac arrest (model of global cerebral ischemia). Our results suggest that PAME can enhance CBF under normal physiological conditions, while administration of PAME (0.02 mg/kg) immediately after cardiopulmonary resuscitation can also enhance CBF in vivo. Additionally, functional learning and spatial memory assessments (via T-maze) 3 days after asphyxial cardiac arrest (ACA) suggest that PAME-treated rats have improved learning and memory recovery versus ACA alone. Furthermore, improved neuronal survival in the CA1 region of the hippocampus were observed in PAME-treated, ACA-induced rats. Altogether, our findings suggest that PAME can enhance CBF, alleviate neuronal cell death, and promote functional outcomes in the presence of ACA.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Parada Cardíaca/prevenção & controle , Fármacos Neuroprotetores/administração & dosagem , Palmitatos/administração & dosagem , Animais , Reanimação Cardiopulmonar , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Parada Cardíaca/terapia , Aprendizagem/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Palmitatos/farmacologia , Ratos , Ratos Sprague-Dawley , Memória Espacial/efeitos dos fármacos
8.
J Vis Exp ; (131)2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29364254

RESUMO

BACKGROUND: Evaluating mild to moderate cognitive impairment in a global cerebral ischemia (i.e. cardiac arrest) model can be difficult due to poor locomotion after surgery. For example, rats who undergo surgical procedures and are subjected to the Morris water maze may not be able to swim, thus voiding the experiment. New Method: We established a modified behavioral spontaneous alternation T-maze test. The major advantage of the modified T-maze protocol is its relatively simple design that is powerful enough to assess functional learning/memory after ischemia. Additionally, the data analysis is simple and straightforward. We used the T-maze to determine the rats' learning/memory deficits both in the presence or absence of mild to moderate (6 min) asphyxial cardiac arrest (ACA). Rats have a natural tendency for exploration and will explore the alternate arms in the T-maze, whereas hippocampal-lesioned rats tend to adopt a side-preference resulting in decreased spontaneous alternation ratios, revealing the hippocampal-related functional learning/memory in the presence or absence of ACA. RESULTS: ACA groups have higher side-preference ratios and lower alternations as compared to control. Comparison with Existing Method(s): The Morris water and Barnes maze are more prominent for assessing learning/memory function. However, the Morris water maze is more stressful than other mazes. The Barnes maze is widely used to measure reference (long-term) memory, while ACA-induced neurocognitive deficits are more closely related to working (short-term) memory. CONCLUSIONS: We have developed a simple, yet effective strategy to delineate working (short-term) memory via the T-maze in our global cerebral ischemia model (ACA).


Assuntos
Parada Cardíaca/fisiopatologia , Transtornos da Memória/diagnóstico , Animais , Parada Cardíaca/diagnóstico , Parada Cardíaca/cirurgia , Masculino , Transtornos da Memória/fisiopatologia , Ratos
9.
Am J Physiol Heart Circ Physiol ; 312(1): H182-H188, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27864234

RESUMO

Sympathetic nervous system activity is increased after cardiopulmonary arrest, resulting in vasoconstrictor release from the perivascular sympathetic nerves of cerebral arteries. However, the pathophysiological function of the perivascular sympathetic nerves in the ischemic brain remains unclear. A rat model of global cerebral ischemia (asphyxial cardiac arrest, ACA) was used to investigate perivascular sympathetic nerves of cerebral arteries via bilateral decentralization (preganglionic lesion) of the superior cervical ganglion (SCG). Decentralization of the SCG 5 days before ACA alleviated hypoperfusion and afforded hippocampal neuroprotection and improved functional outcomes. These studies can provide further insights into the functional mechanism(s) of the sympathetic nervous system during ischemia. NEW & NOTEWORTHY: Interruption of the perivascular sympathetic nerves can alleviate CA-induced hypoperfusion and neuronal cell death in the CA1 region of the hippocampus to enhance functional learning and memory.


Assuntos
Isquemia Encefálica/patologia , Região CA1 Hipocampal/patologia , Artérias Cerebrais/inervação , Neurônios/patologia , Acoplamento Neurovascular , Gânglio Cervical Superior , Simpatectomia , Sistema Nervoso Simpático , Animais , Asfixia/etiologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Isquemia Encefálica/fisiopatologia , Região CA1 Hipocampal/fisiopatologia , Morte Celular , Modelos Animais de Doenças , Parada Cardíaca/complicações , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Microscopia Confocal , Neuroproteção , Ratos , Ratos Sprague-Dawley
10.
Stroke ; 46(8): 2293-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26159789

RESUMO

BACKGROUND AND PURPOSE: Prophylactic treatments that afford neuroprotection against stroke may emerge from the field of preconditioning. Resveratrol mimics ischemic preconditioning, reducing ischemic brain injury when administered 2 days before global ischemia in rats. This protection is linked to silent information regulator 2 homologue 1 (Sirt1) and enhanced mitochondrial function possibly through its repression of uncoupling protein 2. Brain-derived neurotrophic factor (BDNF) is another neuroprotective protein associated with Sirt1. In this study, we sought to identify the conditions of resveratrol preconditioning (RPC) that most robustly induce neuroprotection against focal ischemia in mice. METHODS: We tested 4 different RPC paradigms against a middle cerebral artery occlusion model of stroke. Infarct volume and neurological score were calculated 24 hours after middle cerebral artery occlusion. Sirt1-chromatin binding was evaluated by ChIP-qPCR. Percoll gradients were used to isolate synaptic fractions, and changes in protein expression were determined via Western blot analysis. BDNF concentration was measured using a BDNF-specific ELISA assay. RESULTS: Although repetitive RPC induced neuroprotection from middle cerebral artery occlusion, strikingly one application of RPC 14 days before middle cerebral artery occlusion showed the most robust protection, reducing infarct volume by 33% and improving neurological score by 28%. Fourteen days after RPC, Sirt1 protein was increased 1.5-fold and differentially bound to the uncoupling protein 2 and BDNF promoter regions. Accordingly, synaptic uncoupling protein 2 level decreased by 23% and cortical BDNF concentration increased 26%. CONCLUSIONS: RPC induces a novel extended window of ischemic tolerance in the brain that lasts for at least 14 days. Our data suggest that this tolerance may be mediated by Sirt1 through upregulation of BDNF and downregulation of uncoupling protein 2.


Assuntos
Isquemia Encefálica/prevenção & controle , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Estilbenos/administração & dosagem , Animais , Encéfalo/patologia , Isquemia Encefálica/patologia , Esquema de Medicação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Resveratrol , Fatores de Tempo
11.
PLoS One ; 10(5): e0124918, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25933411

RESUMO

Cardiopulmonary arrest is a leading cause of death and disability in the United States that usually occurs in the aged population. Cardiac arrest (CA) induces global ischemia, disrupting global cerebral circulation that results in ischemic brain injury and leads to cognitive impairments in survivors. Ischemia-induced neuronal damage in the hippocampus following CA can result in the impairment of cognitive function including spatial memory. In the present study, we used a model of asphyxial CA (ACA) in nine month old male Fischer 344 rats to investigate cognitive and synaptic deficits following mild global cerebral ischemia. These experiments were performed with the goals of 1) establishing a model of CA in nine month old middle-aged rats; and 2) to test the hypothesis that learning and memory deficits develop following mild global cerebral ischemia in middle-aged rats. To test this hypothesis, spatial memory assays (Barnes circular platform maze and contextual fear conditioning) and field recordings (long-term potentiation and paired-pulse facilitation) were performed. We show that following ACA in nine month old middle-aged rats, there is significant impairment in spatial memory formation, paired-pulse facilitation n dysfunction, and a reduction in the number of non-compromised hippocampal Cornu Ammonis 1 and subiculum neurons. In conclusion, nine month old animals undergoing cardiac arrest have impaired survival, deficits in spatial memory formation, and synaptic dysfunction.


Assuntos
Transtornos Cognitivos/fisiopatologia , Parada Cardíaca/fisiopatologia , Hipocampo/fisiopatologia , Plasticidade Neuronal , Envelhecimento , Animais , Asfixia/complicações , Asfixia/fisiopatologia , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Transtornos Cognitivos/complicações , Transtornos Cognitivos/patologia , Condicionamento Psicológico , Potenciais Pós-Sinápticos Excitadores , Medo , Reação de Congelamento Cataléptica , Parada Cardíaca/complicações , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Ratos Endogâmicos F344 , Análise de Sobrevida , Transmissão Sináptica
12.
J Cereb Blood Flow Metab ; 35(1): 121-30, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25370861

RESUMO

Ischemic preconditioning (IPC) via protein kinase C epsilon (PKCɛ) activation induces neuroprotection against lethal ischemia. Brain-derived neurotrophic factor (BDNF) is a pro-survival signaling molecule that modulates synaptic plasticity and neurogenesis. Interestingly, BDNF mRNA expression increases after IPC. In this study, we investigated whether IPC or pharmacological preconditioning (PKCɛ activation) promoted BDNF-induced neuroprotection, if neuroprotection by IPC or PKCɛ activation altered neuronal excitability, and whether these changes were BDNF-mediated. We used both in vitro (hippocampal organotypic cultures and cortical neuronal-glial cocultures) and in vivo (acute hippocampal slices 48 hours after preconditioning) models of IPC or PKCɛ activation. BDNF protein expression increased 24 to 48 hours after preconditioning, where inhibition of the BDNF Trk receptors abolished neuroprotection against oxygen and glucose deprivation (OGD) in vitro. In addition, there was a significant decrease in neuronal firing frequency and increase in threshold potential 48 hours after preconditioning in vivo, where this threshold modulation was dependent on BDNF activation of Trk receptors in excitatory cortical neurons. In addition, 48 hours after PKCɛ activation in vivo, the onset of anoxic depolarization during OGD was significantly delayed in hippocampal slices. Overall, these results suggest that after IPC or PKCɛ activation, there are BDNF-dependent electrophysiologic modifications that lead to neuroprotection.


Assuntos
Potenciais de Ação/fisiologia , Isquemia Encefálica/metabolismo , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Hipocampo/metabolismo , Precondicionamento Isquêmico , Proteína Quinase C-épsilon/metabolismo , Animais , Western Blotting , Isquemia Encefálica/patologia , Morte Celular , Técnicas de Cocultura , Ativação Enzimática , Feminino , Hipocampo/patologia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Neuroglia/patologia , Neuroglia/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/patologia , Neurônios/fisiologia , Técnicas de Cultura de Órgãos , Ratos Sprague-Dawley
13.
J Cereb Blood Flow Metab ; 34(6): 1024-32, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24667915

RESUMO

Preserving mitochondrial pools of nicotinamide adenine dinucleotide (NAD) or nicotinamide phosphoribosyltransferase (Nampt), an enzyme involved in NAD production, maintains mitochondrial function and confers neuroprotection after ischemic stress. However, the mechanisms involved in regulating mitochondrial-localized Nampt or NAD have not been defined. In this study, we investigated the roles of protein kinase C epsilon (PKCɛ) and AMP-activated protein kinase (AMPK) in regulating mitochondrial pools of Nampt and NAD after resveratrol or ischemic preconditioning (IPC) in the cortex and in primary neuronal-glial cortical cultures. Using the specific PKCɛ agonist ψɛRACK, we found that PKCɛ induced robust activation of AMPK in vitro and in vivo and that AMPK was required for PKCɛ-mediated ischemic neuroprotection. In purified mitochondrial fractions, PKCɛ enhanced Nampt levels in an AMPK-dependent manner and was required for increased mitochondrial Nampt after IPC or resveratrol treatment. Analysis of intrinsic NAD autofluorescence using two-photon microscopy revealed that PKCɛ modulated NAD in the mitochondrial fraction. Further assessments of mitochondrial NAD concentrations showed that PKCɛ has a key role in regulating the mitochondrial NAD(+)/nicotinamide adenine dinucleotide reduced (NADH) ratio after IPC and resveratrol treatment in an AMPK- and Nampt-dependent manner. These findings indicate that PKCɛ is critical to increase or maintain mitochondrial Nampt and NAD after pathways of ischemic neuroprotection in the brain.


Assuntos
Córtex Cerebral/metabolismo , Citocinas/metabolismo , Inibidores Enzimáticos/farmacologia , Precondicionamento Isquêmico , Mitocôndrias/metabolismo , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Proteína Quinase C-épsilon/metabolismo , Estilbenos/farmacologia , Animais , Células Cultivadas , Ratos , Ratos Sprague-Dawley , Resveratrol
14.
J Cereb Blood Flow Metab ; 34(4): 613-20, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24447953

RESUMO

We previously showed that inhibition of protein kinase C delta (PKCδ) improves brain perfusion 24 hours after asphyxial cardiac arrest (ACA) and confers neuroprotection in the cortex and CA1 region of the hippocampus 7 days after arrest. Therefore, in this study, we investigate the mechanism of action of PKCδ-mediated hypoperfusion after ACA in the rat by using the two-photon laser scanning microscopy (TPLSM) to observe cortical cerebral blood flow (CBF) and laser Doppler flowmetry (LDF) detecting regional CBF in the presence/absence of δV1-1 (specific PKCδ inhibitor), nitric oxide synthase (NOS) substrate (L-arginine, L-arg) and inhibitor (N(ω)-Nitro-L-arginine, NLA), and nitric oxide (NO) donor (sodium nitroprusside, SNP). There was an increase in regional LDF and local (TPLSM) CBF in the presence of δV1-1+L-arg, but only an increase in regional CBF under δV1-1+SNP treatments. Systemic blood nitrite levels were measured 15 minutes and 24 hours after ACA. Nitrite levels were enhanced by pretreatment with δV1-1 30 minutes before ACA possibly attributable to enhanced endothelial NOS protein levels. Our results suggest that PKCδ can modulate NO machinery in cerebral vasculature. Protein kinase C delta can depress endothelial NOS blunting CBF resulting in hypoperfusion, but can be reversed with δV1-1 improving brain perfusion, thus providing subsequent neuroprotection after ACA.


Assuntos
Velocidade do Fluxo Sanguíneo/fisiologia , Córtex Cerebral/irrigação sanguínea , Circulação Cerebrovascular/fisiologia , Parada Cardíaca/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Animais , Arginina/farmacologia , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Circulação Cerebrovascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Parada Cardíaca/fisiopatologia , Fluxometria por Laser-Doppler , Masculino , Microscopia Confocal , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Nitritos/sangue , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Especificidade por Substrato
15.
Transl Stroke Res ; 5(1): 109-17, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24323706

RESUMO

We previously showed that palmitic acid methyl ester (PAME) and stearic acid methyl ester (SAME) are simultaneously released from the sympathetic ganglion and PAME possesses potent vasodilatory properties which may be important in cerebral ischemia. Since PAME is a potent vasodilator simultaneously released with SAME, our hypothesis was that PAME/SAME confers neuroprotection in rat models of focal/global cerebral ischemia. We also examined the neuroprotective properties of Solutol HS15, a clinically approved excipient because it possesses similar fatty acid compositions as PAME/SAME. Asphyxial cardiac arrest (ACA, 6 min) was performed 30 min after PAME/SAME treatment (0.02 mg/kg, IV). Solutol HS15 (2 ml/kg, IP) was injected chronically for 14 days (once daily). Histopathology of hippocampal CA1 neurons was assessed 7 days after ACA. For focal ischemia experiments, PAME, SAME, or Solutol HS15 was administered following reperfusion after 2 h of middle cerebral artery occlusion (MCAO). 2,3,5-Triphenyltetrazolium staining of the brain was performed 24 h after MCAO and the infarct volume was quantified. Following ACA, the number of surviving hippocampal neurons was enhanced by PAME-treated (68%), SAME-treated (69%), and Solutol-treated HS15 (68%) rats as compared to ACA only-treated groups. Infarct volume was decreased by PAME (83%), SAME (68%), and Solutol HS15 (78%) as compared to saline (vehicle) in MCAO-treated animals. PAME, SAME, and Solutol HS15 provide robust neuroprotection in both paradigms of ischemia. This may prove therapeutically beneficial since Solutol HS15 is already administered as a solublizing agent to patients. With proper timing and dosage, administration of Solutol HS15 and PAME/SAME can be an effective therapy against cerebral ischemia.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Palmitatos/uso terapêutico , Polietilenoglicóis/uso terapêutico , Ácidos Esteáricos/uso terapêutico , Vasodilatadores/uso terapêutico , Animais , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/tratamento farmacológico , Masculino , Ratos , Ratos Sprague-Dawley
16.
Curr Drug Targets ; 14(1): 20-35, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23170794

RESUMO

Cardiopulmonary arrest is one of the leading causes of death and disability, primarily occurring in the aged population. Numerous global cerebral ischemia animal models induce neuronal damage similar to cardiac arrest. These global cerebral ischemia models range from vessel occlusion to total cessation of cardiac function, both of which have allowed for the investigation of this multifaceted disease and detection of numerous agents that are neuroprotective. Synapses endure a variety of alterations after global cerebral ischemia from the resulting excitotoxicity and have been a major target for neuroprotection; however, neuroprotective agents have proven unsuccessful in clinical trials, as neurological outcomes have not displayed significant improvements in patients. A majority of these neuroprotective agents have specific neuronal targets, where the success of future neuroprotective agents may depend on non-specific targets and numerous cognitive improvements. This review focuses on the different models of global cerebral ischemia, neuronal synaptic alterations, synaptic neuroprotection and behavioral tests that can be used to determine deficits in cognitive function after global cerebral ischemia.


Assuntos
Isquemia Encefálica/complicações , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Transmissão Sináptica/efeitos dos fármacos , Envelhecimento/psicologia , Animais , Comportamento Animal/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Isquemia Encefálica/psicologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Transtornos Cognitivos/patologia , Transtornos Cognitivos/prevenção & controle , Transtornos Cognitivos/psicologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/uso terapêutico , Transmissão Sináptica/fisiologia
17.
Am J Physiol Cell Physiol ; 303(6): C682-97, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22785120

RESUMO

Coupled gating (synchronous openings and closures) of groups of skeletal muscle ryanodine receptors (RyR1), which mimics RyR1-mediated Ca(2+) release underlying Ca(2+) sparks, was first described by Marx et al. (Marx SO, Ondrias K, Marks AR. Science 281: 818-821, 1998). The nature of the RyR1-RyR1 interactions for coupled gating still needs to be characterized. Consequently, we defined planar lipid bilayer conditions where ∼25% of multichannel reconstitutions contain mixtures of coupled and independently gating RyR1. In ∼10% of the cases, all RyRs (2-10 channels; most frequently 3-4) gated in coupled fashion, allowing for quantification. Our results indicated that coupling required cytosolic solutions containing ATP/Mg(2+) and high (50 mM) luminal Ca(2+) (Ca(lum)) or Sr(2+) solutions. Bursts of coupled activity (events) started and ended abruptly, with all channels activating/deactivating within ∼300 µs. Coupled RyR1 were heterogeneous, where highly active RyR1 ("drivers") seemed open during the entire coupled event (P(o) = 1), while other RyR1s ("followers") displayed abundant flickering and smaller amplitude. Drivers mean open time increased with cytosolic Ca(2+) (Ca(cyt)) or caffeine, whereas followers flicker frequency was Ca(cyt) independent and more sensitive to inhibition by cytosolic Mg(2+). Coupled events were insensitive to varying lumen-to-cytosol Ca(2+) fluxes from ∼1 to 8 pA, which does not corroborate coupling of neighboring RyR1 by local Ca(2+)-induced Ca(2+) release. However, coupling requires specific Ca(lum) sites, as it was lost when Ca(lum) was replaced by luminal Ba(2+) or Mg(2+). In summary, coupled events reveal complex interactions among heterogeneous RyR1, differentially modulated by cytosolic ATP/Mg(2+), Ca(cyt), and Ca(lum,) which under cell-like ionic conditions may parallel synchronous RyR1 gating during Ca(2+) sparks.


Assuntos
Trifosfato de Adenosina/fisiologia , Cálcio/fisiologia , Ativação do Canal Iônico/fisiologia , Magnésio/fisiologia , Músculo Esquelético/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Trifosfato de Adenosina/química , Animais , Cálcio/química , Magnésio/química , Músculo Esquelético/fisiologia , Coelhos , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia
18.
Mol Pharmacol ; 80(3): 509-17, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21665944

RESUMO

Various pharmacological agents designed to modulate plasma membrane ion channels seem to significantly affect intracellular Ca²âº signaling when acting on their target receptor. Some agents could also cross-react (modulate channels or receptors beyond their putative target) with intracellular Ca²âº transporters. This study investigated the potential of thirty putative modulators of either plasma membrane K⁺, Na⁺, or transient receptor potential (TRP) channels to cross-react with intracellular Ca²âº release channels [i.e., ryanodine receptors (RyRs)] from skeletal muscle sarcoplasmic reticulum (SR). Screening for cross-reactivity of these various agents was performed by measuring the rate of spontaneous Ca²âº leak or caffeine-induced Ca²âº release from SR microsomes. Four of the agents displayed a strong cross-reactivity and were further evaluated with skeletal RyR (RyR1) reconstituted into planar bilayers. 6,12,19,20,25,26-Hexahydro-5,27:13,18:21,24-trietheno-11,7-metheno-7H-dibenzo [b,n][1,5,12,16]tetraazacyclotricosine-5, 13-diium dibromide (UCL 1684; K⁺ channel antagonist) and lamotrigine (Na⁺ channel antagonist) were found to significantly inhibit the RyR1-mediated caffeine-induced Ca²âº release. TRP channel agonists anandamide and (-)menthol were found to inhibit and activate RyR1, respectively. High concentrations of nine other agents produced partial inhibition of RyR1-mediated Ca²âº release from SR microsomes. Various pharmacological agents, especially TRP modulators, also inhibited a minor RyR1-independent component of the SR Ca²âº leak. Overall, ∼43% of the agents selected cross-reacted with RyR1-mediated and/or RyR1-independent Ca²âº leak from intracellular stores. Thus, cross-reactivity should be considered when using these classes of pharmacological agents to determine the role of plasmalemmal channels in Ca²âº homeostasis.


Assuntos
Canais Iônicos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Membrana Celular/enzimologia , Reações Cruzadas , Microssomos/enzimologia , Músculo Esquelético/enzimologia , Coelhos
19.
Mol Pharmacol ; 79(1): 141-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20923851

RESUMO

7-Chloro-5-(2-chlorophenyl)-1,5-dihydro-4,1-benzothiazepin-2(3H)-one [CGP-37157 (CGP)], a benzothiazepine derivative of clonazepam, is commonly used as a blocker of the mitochondrial Na+/Ca²+ exchanger. However, evidence suggests that CGP could also affect other targets, such as L-type Ca²+ channels and plasmalemma Na+/Ca²+ exchanger. Here, we tested the possibility of a direct modulation of ryanodine receptor channels (RyRs) and/or sarco/endoplasmic reticulum Ca²+-stimulated ATPase (SERCA) by CGP. In the presence of ruthenium red (inhibitor of RyRs), CGP decreased SERCA-mediated Ca²+ uptake of cardiac and skeletal sarcoplasmic reticulum (SR) microsomes (IC50 values of 6.6 and 9.9 µM, respectively). The CGP effects on SERCA activity correlated with a decreased V(max) of ATPase activity of SERCA-enriched skeletal SR fractions. CGP (≥ 5 µM) also increased RyR-mediated Ca²+ leak from skeletal SR microsomes. Planar bilayer studies confirmed that both cardiac and skeletal RyRs are directly activated by CGP (EC(50) values of 9.4 and 12.0 µM, respectively). In summary, we found that CGP inhibits SERCA and activates RyR channels. Hence, the action of CGP on cellular Ca²+ homeostasis reported in the literature of cardiac, skeletal muscle, and other nonmuscle systems requires further analysis to take into account the contribution of all CGP-sensitive Ca²+ transporters.


Assuntos
ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Clonazepam/análogos & derivados , Músculo Estriado/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Tiazepinas/farmacologia , Animais , Cálcio/metabolismo , Clonazepam/farmacologia , Músculo Estriado/efeitos dos fármacos , Coelhos , Retículo Sarcoplasmático/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...