Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.701
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Int J Med Sci ; 21(7): 1274-1279, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38818467

RESUMO

Objective: Citicoline can be used to reduce acute ischemic stroke injury via venous infusion, however, its protective effects in the brain extracellular space remain largely unknown. Herein, we investigated the brain protective effects of citicoline administered via the brain extracellular space and sought precise effective dosage range that can protect against ischemic injury after experimental ischemic stroke in rats. Methods: Fifty-six Sprague-Dawley rats were randomly divided into control, intraperitoneal (IP), caudate-putamen (CPu)-25, CPu-40, CPu-50, CPu-60 and CPu-75 groups based on the infusion site and concentration of citicoline. Two hours after the administration of citicoline, the rats were subjected to a permanent middle cerebral artery occlusion to mimic acute ischemic stroke. Then, the brain infarct volume in rats after stroke was measured and their neurological deficiency was evaluated to explain the protective effects and effective dosage range of citicoline. Results: Compared to the control and IP groups, brain infarct volume of rats in CPu-40, CPu-50, and CPu-60 groups is significant smaller. Furthermore, the brain infarct volume of rats in CPu-50 is the least. Conclusions: Here, we showed that citicoline can decrease the brain infarct volume, thus protecting the brain from acute ischemic stroke injury. We also found that the appropriate effective citicoline dose delivered via the brain extracellular space is 50 mM. Our study provides novel insights into the precise treatment of acute ischemic stroke by citicoline via the brain extracellular space, further guiding the treatment of brain disease.


Assuntos
Encéfalo , Citidina Difosfato Colina , Modelos Animais de Doenças , Espaço Extracelular , AVC Isquêmico , Ratos Sprague-Dawley , Animais , Citidina Difosfato Colina/administração & dosagem , Citidina Difosfato Colina/farmacologia , Citidina Difosfato Colina/uso terapêutico , Ratos , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/patologia , Espaço Extracelular/efeitos dos fármacos , Masculino , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia
2.
Am J Physiol Cell Physiol ; 322(1): C38-C48, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34788146

RESUMO

The gaseous signaling molecule hydrogen sulfide (H2S) physiologically regulates store-operated Ca2+ entry (SOCE). The SOCE machinery consists of the plasma membrane-localized Orai channels (Orai1-3) and endoplasmic reticulum-localized stromal interaction molecule (STIM)1 and STIM2 proteins. H2S inhibits Orai3- but not Orai1- or Orai2-mediated SOCE. The current objective was to define the mechanism by which H2S selectively modifies Orai3. We measured SOCE and STIM1/Orai3 dynamics and interactions in HEK293 cells exogenously expressing fluorescently tagged human STIM1 and Orai3 in the presence and absence of the H2S donor GYY4137. Two cysteines (C226 and C232) are present in Orai3 that are absent in the Orai1 and Orai2. When we mutated either of these cysteines to serine, alone or in combination, SOCE inhibition by H2S was abolished. We also established that inhibition was dependent on an interaction with STIM1. To further define the effects of H2S on STIM1/Orai3 interaction, we performed a series of fluorescence recovery after photobleaching (FRAP), colocalization, and fluorescence resonance energy transfer (FRET) experiments. Treatment with H2S did not affect the mobility of Orai3 in the membrane, nor did it influence STIM1/Orai3 puncta formation or STIM1-Orai3 protein-protein interactions. These data support a model in which H2S modification of Orai3 at cysteines 226 and 232 limits SOCE evoked upon store depletion and STIM1 engagement, by a mechanism independent of the interaction between Orai3 and STIM1.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Cisteína/metabolismo , Sulfeto de Hidrogênio/toxicidade , Proteínas Sensoras de Cálcio Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Células HEK293 , Humanos , Proteínas Sensoras de Cálcio Intracelular/antagonistas & inibidores , Proteínas de Membrana/antagonistas & inibidores
3.
Proc Natl Acad Sci U S A ; 116(51): 25389-25391, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31792175

RESUMO

Endometriosis is a chronic pain condition affecting ∼176 million women worldwide. It is defined by the presence of endometrium-like tissue (lesions) outside the uterus, most commonly on the pelvic peritoneum. There is no cure for endometriosis. All endometriosis drug approvals to date have been contraceptive, limiting their use in women of child-bearing age. We have shown that human peritoneal mesothelial cells (HPMCs) recovered from the pelvic peritoneum of women with endometriosis exhibit significantly higher glycolysis, lower mitochondrial respiration, decreased enzymatic activity of pyruvate dehydrogenase (PDH), and increased production of lactate compared to HPMCs from women without disease. Transforming growth factor-ß1 (TGF-ß1) is elevated in the peritoneal fluid from women with endometriosis, and exposure of HPMCs to TGF-ß1 exacerbates this abnormal phenotype. Treatment of endometriosis HPMCs with the pyruvate dehydrogenase kinase (PDK) inhibitor/PDH activator dichloroacetate (DCA) normalizes HPMC metabolism, reduces lactate secretion, and abrogates endometrial stromal cell proliferation in a coculture model. Oral DCA reduced peritoneal fluid lactate concentrations and endometriosis lesion size in a mouse model. These findings provide the rationale for targeting metabolic processes as a noncontraceptive treatment for women with endometriosis either as a primary nonhormonal treatment or to prevent recurrence after surgery.


Assuntos
Ácido Dicloroacético/farmacologia , Reposicionamento de Medicamentos , Endometriose , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Espaço Extracelular/efeitos dos fármacos , Feminino , Glicólise/efeitos dos fármacos , Humanos , Camundongos , Peritônio/citologia
4.
J Neurosci ; 40(49): 9440-9454, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33122389

RESUMO

Understanding the effects of anesthesia on cortical neuronal spiking and information transfer could help illuminate the neuronal basis of the conscious state. Recent investigations suggest that the brain state identified by local field potential spectrum is not stationary but changes spontaneously at a fixed level of anesthetic concentration. How cortical unit activity changes with dynamically transitioning brain states under anesthesia is unclear. Extracellular unit activity was measured with 64-channel silicon microelectrode arrays in cortical layers 5/6 of the primary visual cortex of chronically instrumented, freely moving male rats (n = 7) during stepwise reduction of the anesthetic desflurane (6%, 4%, 2%, and 0%). Unsupervised machine learning applied to multiunit spike patterns revealed five distinct brain states. A novel desynchronized brain state with increased spike rate variability, sample entropy, and EMG activity occurred in 6% desflurane with 40.0% frequency. The other four brain states reflected graded levels of anesthesia. As anesthesia deepened the spike rate of neurons decreased regardless of their spike rate profile at baseline conscious state. Actively firing neurons with wide-spiking pattern showed increased bursting activity along with increased spike timing variability, unit-to-population correlation, and unit-to-unit transfer entropy, despite the overall decrease in transfer entropy. The narrow-spiking neurons showed similar changes but to a lesser degree. These results suggest that (1) anesthetic effect on spike rate is distinct from sleep, (2) synchronously fragmented spiking pattern is a signature of anesthetic-induced unconsciousness, and (3) the paradoxical, desynchronized brain state in deep anesthesia contends the generally presumed monotonic, dose-dependent anesthetic effect on the brain.SIGNIFICANCE STATEMENT Recent studies suggest that spontaneous changes in brain state occur under anesthesia. However, the spiking behavior of cortical neurons associated with such state changes has not been investigated. We found that local brain states defined by multiunit activity had a nonunitary relationship with the current anesthetic level. A paradoxical brain state displaying asynchronous firing pattern and high EMG activity was found unexpectedly in deep anesthesia. In contrast, the synchronous fragmentation of neuronal spiking appeared to be a robust signature of the state of anesthesia. The findings challenge the assumption of monotonic, anesthetic dose-dependent behavior of cortical neuron populations. They enhance the interpretation of neuroscientific data obtained under anesthesia and the understanding of the neuronal basis of anesthetic-induced state of unconsciousness.


Assuntos
Anestesia , Encéfalo/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Anestésicos Inalatórios/farmacologia , Animais , Desflurano/farmacologia , Relação Dose-Resposta a Droga , Eletrodos Implantados , Eletroencefalografia , Eletromiografia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Entropia , Espaço Extracelular/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Sono/fisiologia , Inconsciência/induzido quimicamente , Inconsciência/fisiopatologia
5.
J Neurosci ; 40(27): 5161-5176, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32444385

RESUMO

Alterations of excitatory synaptic function are the strongest correlate to the pathologic disturbance of cognitive ability observed in the early stages of Alzheimer's disease (AD). This pathologic feature is driven by amyloid-ß oligomers (Aßos) and propagates from neuron to neuron. Here, we investigated the mechanism by which Aßos affect the function of synapses and how these alterations propagate to surrounding healthy neurons. We used complementary techniques ranging from electrophysiological recordings and molecular biology to confocal microscopy in primary cortical cultures, and from acute hippocampal and cortical slices from male wild-type and amyloid precursor protein (APP) knock-out (KO) mice to assess the effects of Aßos on glutamatergic transmission, synaptic plasticity, and dendritic spine structure. We showed that extracellular application of Aßos reduced glutamatergic synaptic transmission and long-term potentiation. These alterations were not observed in APP KO neurons, suggesting that APP expression is required. We demonstrated that Aßos/APP interaction increases the amyloidogenic processing of APP leading to intracellular accumulation of newly produced Aßos. Intracellular Aßos participate in synaptic dysfunctions as shown by pharmacological inhibition of APP processing or by intraneuronal infusion of an antibody raised against Aßos. Furthermore, we provide evidence that following APP processing, extracellular release of Aßos mediates the propagation of the synaptic pathology characterized by a decreased spine density of neighboring healthy neurons in an APP-dependent manner. Together, our data unveil a complementary role for Aßos in AD, while intracellular Aßos alter synaptic function, extracellular Aßos promote a vicious cycle that propagates synaptic pathology from diseased to healthy neurons.SIGNIFICANCE STATEMENT Here we provide the proof that a vicious cycle between extracellular and intracellular pools of Aß oligomers (Aßos) is required for the spreading of Alzheimer's disease (AD) pathology. We showed that extracellular Aßos propagate excitatory synaptic alterations by promoting amyloid precursor protein (APP) processing. Our results also suggest that subsequent to APP cleavage two pools of Aßos are produced. One pool accumulates inside the cytosol, inducing the loss of synaptic plasticity potential. The other pool is released into the extracellular space and contributes to the propagation of the pathology from diseased to healthy neurons. Pharmacological strategies targeting the proteolytic cleavage of APP disrupt the relationship between extracellular and intracellular Aß, providing a therapeutic approach for the disease.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/metabolismo , Sinapses/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Animais , Anticorpos Bloqueadores/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Histidina/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Cultura Primária de Células , Transmissão Sináptica/efeitos dos fármacos
6.
Biochem Biophys Res Commun ; 546: 138-144, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33582556

RESUMO

Hepatic injury is common in patients who suffer from severe burns plus delayed resuscitation (B + DR). Stimulator of interferon genes (STING) is primarily expressed in Kupffer cells (KCs). We demonstrated that B + DR caused hepatic injury and oxidative stress. Reactive oxygen species (ROS) damage mitochondrial membranes in hepatocytes, leading to the release of mitochondrial DNA (mtDNA) into the hepatocyte cytosol and the circulation. The damaged hepatocytes then activate the mtDNA/STING pathway in KCs and trigger KCs polarization towards pro-inflammatory phenotype. SS-31 is a strong antioxidant that specifically concentrates in the inner mitochondrial membrane. SS-31 prevented hepatic injury by neutralizing ROS, inhibiting the release of mtDNA, protecting hepatocyte mitochondria, suppressing the activation of the mtDNA/STING pathway and inhibiting KCs polarization into pro-inflammatory phenotype.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Queimaduras/complicações , DNA Mitocondrial/efeitos dos fármacos , Células de Kupffer/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/lesões , Proteínas de Membrana/metabolismo , Oligopeptídeos/farmacologia , Ressuscitação , Animais , DNA Mitocondrial/sangue , DNA Mitocondrial/metabolismo , Espaço Extracelular/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Células de Kupffer/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
7.
Circ Res ; 125(9): 787-801, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31434553

RESUMO

RATIONALE: Mechanistic insight into the inflammatory response after acute myocardial infarction may inform new molecularly targeted treatment strategies to prevent chronic heart failure. OBJECTIVE: We identified the sulfatase SULF2 in an in silico secretome analysis in bone marrow cells from patients with acute myocardial infarction and detected increased sulfatase activity in myocardial autopsy samples. SULF2 (Sulf2 in mice) and its isoform SULF1 (Sulf1) act as endosulfatases removing 6-O-sulfate groups from heparan sulfate (HS) in the extracellular space, thus eliminating docking sites for HS-binding proteins. We hypothesized that the Sulfs have a role in tissue repair after myocardial infarction. METHODS AND RESULTS: Both Sulfs were dynamically upregulated after coronary artery ligation in mice, attaining peak expression and activity levels during the first week after injury. Sulf2 was expressed by monocytes and macrophages, Sulf1 by endothelial cells and fibroblasts. Infarct border zone capillarization was impaired, scar size increased, and cardiac dysfunction more pronounced in mice with a genetic deletion of either Sulf1 or Sulf2. Studies in bone marrow-chimeric Sulf-deficient mice and Sulf-deficient cardiac endothelial cells established that inflammatory cell-derived Sulf2 and endothelial cell-autonomous Sulf1 promote angiogenesis. Mechanistically, both Sulfs reduced HS sulfation in the infarcted myocardium, thereby diminishing Vegfa (vascular endothelial growth factor A) interaction with HS. Along this line, both Sulfs rendered infarcted mouse heart explants responsive to the angiogenic effects of HS-binding Vegfa164 but did not modulate the angiogenic effects of non-HS-binding Vegfa120. Treating wild-type mice systemically with the small molecule HS-antagonist surfen (bis-2-methyl-4-amino-quinolyl-6-carbamide, 1 mg/kg/day) for 7 days after myocardial infarction released Vegfa from HS, enhanced infarct border-zone capillarization, and exerted sustained beneficial effects on cardiac function and survival. CONCLUSIONS: These findings establish HS-editing Sulfs as critical inducers of postinfarction angiogenesis and identify HS sulfation as a therapeutic target for ischemic tissue repair.


Assuntos
Espaço Extracelular/metabolismo , Isquemia Miocárdica/metabolismo , Sulfatases/biossíntese , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Disponibilidade Biológica , Espaço Extracelular/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isquemia Miocárdica/patologia , Fator A de Crescimento do Endotélio Vascular/administração & dosagem
8.
Biol Pharm Bull ; 44(2): 266-270, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33518679

RESUMO

Extracellular pH (pHe) of tumor cells is characteristic of tumor microenvironment (TME). Acidic TME impairs the responses of tumors to some anti-cancer chemotherapies. In this study, we showed that daily oral dosing of sodium potassium citrate (K/Na citrate) increased blood HCO3- concentrations, corresponding to increase of HCO3- concentrations and pHs in urine, and neutralized the tumor pHe. Neutralization of acidic TME by alkaline substance like HCO3-, an active metabolite of K/Na citrate, well potentiated the therapeutic effect of anticancer agent TS-1®, an orally active 5-fuluoro-uracil derivative, in Panc-1 pancreatic cancer-xenograft murine model. Neutralization of acidic TME by using an alkaline K/Na citrate is a smart approach for enhancement of the therapeutic effects of anticancer agents for pancreatic cancer in the end stage.


Assuntos
Antiácidos/administração & dosagem , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Ácido Oxônico/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Tegafur/administração & dosagem , Microambiente Tumoral/efeitos dos fármacos , Administração Oral , Animais , Antiácidos/farmacocinética , Linhagem Celular Tumoral , Combinação de Medicamentos , Sinergismo Farmacológico , Espaço Extracelular/química , Espaço Extracelular/efeitos dos fármacos , Feminino , Humanos , Camundongos , Ácido Oxônico/farmacocinética , Ácido Oxônico/uso terapêutico , Neoplasias Pancreáticas/patologia , Citrato de Potássio/administração & dosagem , Citrato de Potássio/farmacocinética , Citrato de Sódio/administração & dosagem , Citrato de Sódio/farmacocinética , Tegafur/farmacocinética , Tegafur/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Int J Med Sci ; 18(3): 652-659, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33437200

RESUMO

Objective: Different anesthetics have distinct effects on the interstitial fluid (ISF) drainage in the extracellular space (ECS) of the superficial rat brain, while their effects on ISF drainage in the ECS of the deep rat brain still remain unknown. Herein, we attempt to investigate and compare the effects of propofol and isoflurane on ECS structure and ISF drainage in the caudate-putamen (CPu) and thalamus (Tha) of the deep rat brain. Methods: Adult Sprague-Dawley rats were anesthetized with propofol or isoflurane, respectively. Twenty-four anesthetized rats were randomly divided into the propofol-CPu, isoflurane-CPu, propofol-Tha, and isoflurane-Tha groups. Tracer-based magnetic resonance imaging (MRI) and fluorescent-labeled tracer assay were utilized to quantify ISF drainage in the deep brain. Results: The half-life of ISF in the propofol-CPu and propofol-Tha groups was shorter than that in the isoflurane-CPu and isoflurane-Tha groups, respectively. The ECS volume fraction in the propofol-CPu and propofol-Tha groups was much higher than that in the isoflurane-CPu and isoflurane-Tha groups, respectively. However, the ECS tortuosity in the propofol-CPu and propofol-Tha groups was much smaller than that in isoflurane-CPu and isoflurane-Tha groups, respectively. Conclusions: Our results demonstrate that propofol rather than isoflurane accelerates the ISF drainage in the deep rat brain, which provides novel insights into the selective control of ISF drainage and guides selection of anesthetic agents in different clinical settings, and unravels the mechanism of how general anesthetics function.


Assuntos
Anestésicos Gerais/administração & dosagem , Núcleo Caudado/efeitos dos fármacos , Líquido Extracelular/metabolismo , Putamen/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Administração por Inalação , Animais , Núcleo Caudado/citologia , Núcleo Caudado/diagnóstico por imagem , Núcleo Caudado/metabolismo , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Gadolínio DTPA/administração & dosagem , Infusões Parenterais , Isoflurano/administração & dosagem , Imageamento por Ressonância Magnética/métodos , Modelos Animais , Propofol/administração & dosagem , Putamen/citologia , Putamen/diagnóstico por imagem , Putamen/metabolismo , Ratos , Ratos Sprague-Dawley , Tálamo/citologia , Tálamo/diagnóstico por imagem , Tálamo/metabolismo
10.
J Biol Chem ; 294(9): 3081-3090, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30563841

RESUMO

Hepatocyte lipotoxicity is characterized by aberrant mitochondrial metabolism, which predisposes cells to oxidative stress and apoptosis. Previously, we reported that translocation of calcium from the endoplasmic reticulum to mitochondria of palmitate-treated hepatocytes activates anaplerotic flux from glutamine to α-ketoglutarate (αKG), which subsequently enters the citric acid cycle (CAC) for oxidation. We hypothesized that increased glutamine anaplerosis fuels elevations in CAC flux and oxidative stress following palmitate treatment. To test this hypothesis, primary rat hepatocytes or immortalized H4IIEC3 rat hepatoma cells were treated with lipotoxic levels of palmitate while modulating anaplerotic pathways leading to αKG. We found that culture media supplemented with glutamine, glutamate, or dimethyl-αKG increased palmitate lipotoxicity compared with media that lacked these anaplerotic substrates. Knockdown of glutamate-oxaloacetate transaminase activity significantly reduced the lipotoxic effects of palmitate, whereas knockdown of glutamate dehydrogenase (Glud1) had no effect on palmitate lipotoxicity. 13C flux analysis of H4IIEC3 cells co-treated with palmitate and the pan-transaminase inhibitor aminooxyacetic acid confirmed that reductions in lipotoxic markers were associated with decreases in anaplerosis, CAC flux, and oxygen consumption. Taken together, these results demonstrate that lipotoxic palmitate treatments enhance anaplerosis in cultured rat hepatocytes, causing a shift to aberrant transaminase metabolism that fuels CAC dysregulation and oxidative stress.


Assuntos
Aspartato Aminotransferases/metabolismo , Ciclo do Ácido Cítrico/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Palmitatos/toxicidade , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Glutamina/metabolismo , Hepatócitos/citologia , Ácidos Cetoglutáricos/metabolismo , Masculino , Estresse Oxidativo/efeitos dos fármacos , Oxigênio/metabolismo , Ratos , Ratos Sprague-Dawley
11.
Biol Pharm Bull ; 43(9): 1306-1314, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32879204

RESUMO

The extracellular space (ECS) is the space between the neurons and the capillaries in the brain. The volume fraction (α) and the tortuosity (λ) are the main parameters used to describe its characteristics. Thymoquinone has been proved to possess anti-oxidant and anti-inflammatory activity. In this study, we used a gadolinium-diethylenetriaminepentacetate (Gd-DTPA)-enhanced magnetic resonance imaging (MRI) system to determine the effects of thymoquinone on ECS parameters in transient middle cerebral artery occlusion rats (tMCAO) to prove the neuroprotective effect of thymoquinone on brain tissue damage caused by ischemic stroke. Neurological examinations, 2,3,5-triphenyltetrazolium chloride (TTC) staining, hematoxylin-eosin (H&E) staining and assaying of ECS parameters using MRI were performed 24 h after surgery. We found that thymoquinone could improve the behavioural performance by neurological examinations. TTC staining indicated that thymoquinone significantly decreased the percentage of hemi-cerebral infarction. Also, H&E staining showed that thymoquinone could inhibit the neuron necrosis in the hippocampal CA1 region. We found that thymoquinone treatment could inhibit the changes in ECS diffusion parameters, which might prove that thymoquinone might protect brain tissue damage caused by ischemic stroke. Thymoquinone can protect the brain against cerebral ischemia-reperfusion injury, effectively ameliorate abnormalities in characteristics of ECS and decrease cerebral infarction in tMCAO rats.


Assuntos
Benzoquinonas/administração & dosagem , Encéfalo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , AVC Isquêmico/tratamento farmacológico , Fármacos Neuroprotetores/administração & dosagem , Animais , Encéfalo/citologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Modelos Animais de Doenças , Espaço Extracelular/diagnóstico por imagem , Espaço Extracelular/efeitos dos fármacos , Humanos , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/diagnóstico , Infarto da Artéria Cerebral Média/patologia , AVC Isquêmico/diagnóstico , AVC Isquêmico/etiologia , AVC Isquêmico/patologia , Imageamento por Ressonância Magnética , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos , Ratos Sprague-Dawley
12.
Molecules ; 25(16)2020 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-32781695

RESUMO

Previous studies have reported that estrogen hormone promotes melanogenesis while progesterone inhibits it. A selective estrogen receptor modulator (SERM), tamoxifen, has been shown to promote melanogenesis; however, to date, there have been no reports on the effects of a selective progesterone receptor modulator (SPRM) on melanogenesis. In the present study, we hypothesized that asoprisnil (AP), a SPRM, inhibits melanogenesis. AP was tested for cytotoxicity to B16F10 mouse melanoma cells for screening the nontoxic concentrations using MTS cytotoxicity assay. Extracellular and intracellular melanin levels were estimated at nontoxic concentrations of AP. To evaluate the direct effect of AP on tyrosinase enzyme, tyrosinase activity and copper chelating activities were measured. Next, the effects of AP on melanogenesis were tested in normal human melanocytes, neonatal, darkly pigmented (HEMn-DP). Our results demonstrate that AP was nontoxic at a concentration range of 10-50 µM in B16F10 cells; AP at 50 µM significantly suppressed extracellular melanin levels comparable to kojic acid at 500 µM, with no significant effect on intracellular melanin levels. The mechanism of melanogenesis inhibition was studied to assess if AP downregulated tyrosinase activity in cell lysates or in a cell-free system. However, AP was found to increase intracellular tyrosinase activity without any effect on tyrosinase enzyme activity or copper chelating activity in a cell-free system, indicating that AP inhibits melanogenesis by mechanisms other than direct effects on tyrosinase enzyme activity. The capacity of AP to inhibit melanosome export was further validated in HEMn-DP cells; AP significantly suppressed dendricity at concentrations of 20 and 30 µM in the absence of effects on melanin synthesis or intracellular tyrosinase activity. In addition, AP was nontoxic to human keratinocytes (HaCaT) at these concentrations, validating its safety for topical use. Taken together, our preliminary results demonstrate that AP might be repurposed as a candidate therapeutic for treatment of hyperpigmentation disorders via a unique mechanism, which encompasses a selective inhibition of melanosome export.


Assuntos
Estrenos/farmacologia , Melanossomas/efeitos dos fármacos , Melanossomas/metabolismo , Oximas/farmacologia , Receptores de Progesterona/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Melanoma Experimental/patologia , Camundongos
13.
Biophys J ; 117(10): 1783-1794, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31542225

RESUMO

An improved version of the integrative optical imaging method has been developed that substantially increases the time resolution of diffusion measurements. We present a theory for time-resolved integrative optical imaging that incorporates a time-dependent effective diffusion coefficient in homogeneous anisotropic media and a time-dependent nonspecific linear clearance. The method was applied to measure the very fast changes in extracellular diffusion that occur during spreading depression in rat hippocampal slices. We were able to achieve a time resolution of approximately 1 s, an improvement of at least 10 times compared to the standard methods for extracellular diffusion measurement. We have found that diffusion of a small fluorescent extracellular marker (MW 3000) completely stopped during the maximum direct current shift associated with the spreading depression wave, then gradually resumed over several minutes afterward. The effect of spreading depression on extracellular space is much larger than previously estimated by other methods with lower time resolution.


Assuntos
Potenciais de Ação/fisiologia , Imagem Óptica , Animais , Sistemas Computacionais , Dextranos/metabolismo , Difusão , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Feminino , Iontoforese , Propionatos/farmacologia , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Fatores de Tempo
14.
J Cell Physiol ; 234(6): 9764-9777, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30387146

RESUMO

The migration of endothelial cells (ECs) is closely associated with a Ca2+ -dependent protein, protein kinase Cα (PKCα). The disruption of intercellular adhesion by single-cell wounding has been shown to induce the directional translocation of PKCα. We hypothesized that this translocation of PKCα is induced by mechanical stress, such as unloading of intercellular tension, or by intercellular communication, such as gap junction-mediated and paracrine signaling. In the current study, we found that the disruption of intercellular adhesion induced the directional translocation of PKCα even when gap junction-mediated and paracrine signaling were inhibited. Conversely, it did not occur when the mechanosensitive channel was inhibited. In addition, the strain field of substrate attributable to the disruption of intercellular adhesion tended to be larger at the areas corresponding with PKCα translocation. Recently, we found that a direct mechanical stimulus induced the accumulation of PKCα at the stimulus area, involving Ca 2+ influx from extracellular space. These results indicated that the unloading of intercellular tension induced directional translocation of PKCα, which required Ca 2+ influx from extracellular space. The results of this study indicate the involvement of PKCα in the Ca 2+ signaling pathway in response to mechanical stress in ECs.


Assuntos
Espaço Extracelular/metabolismo , Proteína Quinase C-alfa/metabolismo , Animais , Fenômenos Biomecânicos , Cálcio/metabolismo , Carbazóis/farmacologia , Bovinos , Adesão Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Cinética , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Tapsigargina/farmacologia
15.
J Neurochem ; 151(6): 749-763, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31478210

RESUMO

A significant comorbidity exists between alcohol and methamphetamine (Meth) abuse but the neurochemical consequences of this co-abuse are unknown. Alcohol and Meth independently and differentially affect glutamatergic transmission but the unique effects of their serial exposure on glutamate signaling in mediating damage to dopamine neurons are unknown. Sprague-Dawley rats had intermittent voluntary access to 10% ethanol (EtOH) every other day and water over 28 days and were then administered a binge injection regimen of Meth or saline. EtOH drinking decreased the glutamate aspartate transporter and increased basal extracellular concentrations of glutamate within the striatum when measured after the last day of drinking. Ceftriaxone is known to increase the expression and/or activity of glutamate transporters in the brain and prevented both the decreases in glutamate aspartate transporter and the increases in basal extracellular glutamate when administered during EtOH drinking. EtOH drinking also exacerbated the acute increases in extracellular glutamate observed upon Meth exposure, the subsequent increases in spectrin proteolysis, and the long-term decreases in dopamine content in the striatum, all of which were attenuated by ceftriaxone administration during EtOH drinking only. These results implicate EtOH-induced increases in extracellular glutamate and corresponding decreases in glutamate uptake as mechanisms that contribute to the vulnerability produced by EtOH drinking and the unique neurotoxicity observed after serial exposure to Meth that is not observed with either drug alone. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.


Assuntos
Consumo de Bebidas Alcoólicas/efeitos adversos , Etanol/toxicidade , Ácido Glutâmico/toxicidade , Metanfetamina/toxicidade , Consumo de Bebidas Alcoólicas/metabolismo , Animais , Sinergismo Farmacológico , Etanol/administração & dosagem , Transportador 1 de Aminoácido Excitatório/metabolismo , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Ácido Glutâmico/metabolismo , Masculino , Metanfetamina/administração & dosagem , Microdiálise/métodos , Ratos , Ratos Sprague-Dawley
16.
J Pharmacol Exp Ther ; 370(3): 427-435, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31197021

RESUMO

The Ca2+-sensing receptor (CaSR) detects small changes in extracellular calcium (Ca2+ e) concentration ([Ca2+]e) and transduces the signal into modulation of various signaling pathways. Ca2+-induced relaxation of isolated phenylephrine-contracted mesenteric arteries is mediated by the CaSR of the perivascular nerve. Elucidation of the regulatory mechanisms involved in vascular CaSR signaling may provide insights into the physiologic functions of the receptor and identify targets for the development of new treatments for cardiovascular pathologies such as hypertension. Protein kinase Cα (PKCα) is a critical regulator of multiple signaling pathways and can phosphorylate the CaSR leading to receptor desensitization. In this study, we used automated wire myography to investigate the effects of CaSR mutation and small-interfering RNA downregulation of PKCα on CaSR-mediated relaxation of phenylephrine-contracted mesenteric arteries from aged Dahl salt-sensitive (SS) rats on a low-salt diet. The data showed minimal relaxation responses of arteries to Ca2+ e in wild-type (SS) and CaSR mutant (SS-Casrem1Mcwi) rats. Mutation of the CaSR gene had no significant effect on relaxation. PKCα expression was similar in wild-type and mutant rats, and small-interfering RNA downregulation of PKCα and/or inhibition of PKC with the Ca2+-sensitive GÓ§ 6976 resulted in a >80% increase in relaxation. Significant differences in EC50 values were observed between treated and untreated controls (P < 0.05 analysis of variance). The results indicate that PKCα plays an important role in the regulation of CaSR-mediated relaxation of mesenteric arteries, and its downregulation or pharmacological inhibition may lead to an increased Ca2+ sensitivity of the receptor and reversal of age-related changes in vascular tone. SIGNIFICANCE STATEMENT: G protein-coupled CaSR signaling leads to the regulation of vascular tone and may, therefore, play a vital role in blood pressure regulation. The receptor has several PKC phosphorylation sites in the C-terminal intracellular tail that mediate desensitization. We have previously shown that activation of the CaSR in neuronal cells leads to PKC phosphorylation, indicating that protein kinase C is an important regulator of CaSR function. Therefore, PKC in the CaSR signaling pathway in mesenteric arteries is a potential target for the development of new therapeutic approaches to treat hypertension and age-related vascular dysfunction. The present studies show that small-interfering RNA downregulation of PKCα and pharmacological inhibition of PKC enhanced CaSR-mediated relaxation of phenylephrine-contracted mesenteric arteries from aged Dahl salt-sensitive rats.


Assuntos
Cálcio/metabolismo , Regulação para Baixo , Espaço Extracelular/metabolismo , Artérias Mesentéricas/citologia , Artérias Mesentéricas/fisiologia , Proteína Quinase C/metabolismo , Vasodilatação , Animais , Espaço Extracelular/efeitos dos fármacos , Artérias Mesentéricas/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Endogâmicos Dahl , Receptores de Detecção de Cálcio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
17.
Int J Neuropsychopharmacol ; 22(7): 453-465, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31188434

RESUMO

BACKGROUND: Synapsins are encoded by SYN I, SYN II, and SYN III, and they regulate neurotransmitter release by maintaining a reserve pool of synaptic vesicles. METHODS: Presynaptic dopamine responses to cocaine were examined by microdialysis, and postsynaptic responses were evaluated to various dopamine receptor agonists in the open field with SynI/SynII/SynIII triple knockout mice. RESULTS: Triple knockout mice showed enhanced spontaneous locomotion in a novel environment and were hyper-responsive to indirect and direct D1 and D2 dopamine agonists. Triple knockout animals appeared sensitized to cocaine upon first open field exposure; sensitization developed across days in wild-type controls. When mutants were preexposed to a novel environment before injection, cocaine-stimulated locomotion was reduced and behavioral sensitization retarded. Baseline dopamine turnover was enhanced in mutants and novel open field exposure increased their striatal dopamine synthesis rates. As KCl-depolarization stimulated comparable dopamine release in both genotypes, their readily releasable pools appeared indistinguishable. Similarly, cocaine-induced hyperlocomotion was indifferent to blockade of newly synthesized dopamine and depletion of releasable dopamine pools. Extracellular dopamine release was similar in wild-type and triple knockout mice preexposed to the open field and given cocaine or placed immediately into the arena following injection. Since motor effects to novelty and psychostimulants depend upon frontocortical-striatal inputs, we inhibited triple knockout medial frontal cortex with GABA agonists. Locomotion was transiently increased in cocaine-injected mutants, while their supersensitive cocaine response to novelty was lost. CONCLUSIONS: These results reveal presynaptic dopamine release is not indicative of agonist-induced triple knockout hyperlocomotion. Instead, their novelty response occurs primarily through postsynaptic mechanisms and network effects.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Agonistas de Dopamina/farmacologia , Dopamina/metabolismo , Atividade Motora/efeitos dos fármacos , Sinapses/metabolismo , Sinapsinas/deficiência , Animais , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Feminino , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Agonistas GABAérgicos/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microdiálise , Atividade Motora/fisiologia , Sinapsinas/genética
18.
Cell Mol Biol (Noisy-le-grand) ; 65(5): 73-78, 2019 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-31304910

RESUMO

Heweijiangni decoction (HWJND) is an effective traditional Chinese medicine prescription in clinical treatment of nonerosive reflux disease (NERD). Esophageal hypersensitivity and acid contribute to the disease. However, the exact underlying mechanism of action remains unclear. In this study, we observed the effect of HWJND on esophageal morphology in a rat model of ovalbumin (OVA)-induced visceral hypersensitivity followed by acid exposure. Esophageal morphology was assessed by measuring the extent of dilated intercellular spaces (DIS), desmosome disruption, and mitochondrial fragmentation. HWJND in low, moderate, and high doses relieved DIS and desmosome disruption in esophageal epithelium compared with model group (P<0.05 for all doses). In addition, HWJND in high dose protected mitochondria from fragmentation (P<0.05). Other findings suggest that DIS and mitochondrial fragmentation are independent events, and that omeprazole protects mitochondria. Overall, HWJND significantly resists esophageal morphology changes in OVA-induced and acid exposure rat model.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Esôfago/efeitos dos fármacos , Refluxo Gastroesofágico/induzido quimicamente , Refluxo Gastroesofágico/tratamento farmacológico , Ácido Clorídrico/farmacologia , Ovalbumina/farmacologia , Animais , Desmossomos/efeitos dos fármacos , Modelos Animais de Doenças , Esôfago/patologia , Espaço Extracelular/efeitos dos fármacos , Ácido Clorídrico/administração & dosagem , Injeções Intraperitoneais , Masculino , Mitocôndrias/efeitos dos fármacos , Omeprazol/farmacologia , Ovalbumina/administração & dosagem , Ratos , Ratos Sprague-Dawley
19.
Clin Exp Pharmacol Physiol ; 46(1): 56-64, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30203559

RESUMO

Inflammation elevates intracellular calcium concentrations ([Ca2+ ]i ) in airway smooth muscle (ASM). The L-type Ca2+ channel (L-VDCC) plays an important role in regulating Ca2+ influx in ASM. However, the role of L-VDCC in the inflammatory cytokine-induced pathology of ASM remains unclear. In the present study, we used calcium imaging and isometric tension measurements to assess the role of L-VDCC in agonist-induced [Ca2+ ]i rise and the associated contractions in mouse ASM, and we used immunoblotting to identify L-VDCC protein expression levels in mouse ASM after exposure to tumour necrosis factor alpha (TNF-α) or interleukin-8 (IL-8). Our results showed that high-K+ - or carbachol-induced contractions of mouse ASM were significantly greater after pretreatment with TNF-α or IL-8 for 24 hours. Both verapamil and nifedipine, L-VDCC inhibitors, abolished this increased contraction induced by TNF-α or IL-8 pretreatment. Moreover, TNF-α treatment enhanced carbachol-induced Ca2+ influx in ASM cells, and this effect was abrogated by verapamil. Additionally, immunoblotting results showed that preincubation of mouse ASM with TNF-α or IL-8 also enhanced L-VDCC protein expression. On the basis of these findings, we concluded that proinflammatory cytokines, such as TNF-α and IL-8, increase the expression level of L-VDCC, which in turn contributes to augmented agonist-induced ASM contractions. This effect of inflammation on L-VDCC expression in ASM may be associated with airway hyper-responsiveness and involved in the development of asthma.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-8/farmacologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Traqueia/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Cálcio/metabolismo , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Inflamação/metabolismo , Masculino , Camundongos , Músculo Liso/citologia , Músculo Liso/fisiologia , Nifedipino/farmacologia , Potássio/farmacologia , Traqueia/citologia , Traqueia/efeitos dos fármacos , Verapamil/farmacologia
20.
J Neurochem ; 144(4): 408-420, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29164598

RESUMO

Vitamin C (in the reduced form ascorbate or in the oxidized form dehydroascorbate) is implicated in signaling events throughout the central nervous system (CNS). In the retina, a high-affinity transport system for ascorbate has been described and glutamatergic signaling has been reported to control ascorbate release. Here, we investigated the modulatory role played by vitamin C upon glutamate uptake and N-methyl-d-aspartate (NMDA) receptor activation in cultured retinal cells or in intact retinal tissue using biochemical and imaging techniques. We show that both forms of vitamin C, ascorbate or dehydroascorbate, promote an accumulation of extracellular glutamate by a mechanism involving the inhibition of glutamate uptake. This inhibition correlates with the finding that ascorbate promotes a decrease in cell surface levels of the neuronal glutamate transporter excitatory amino acid transporter 3 in retinal neuronal cultures. Interestingly, vitamin C is prone to increase the activity of NMDA receptors but also promotes a decrease in glutamate-stimulated [3 H] MK801 binding and decreases cell membrane content of NMDA receptor glutamate ionotropic receptor subunit 1 (GluN1) subunits. Both compounds were also able to increase cAMP response element-binding protein phosphorylation in neuronal nuclei in a glutamate receptor and calcium/calmodulin kinase-dependent manner. Moreover, the effect of ascorbate is not blocked by sulfinpyrazone and then does not depend on its uptake by retinal cells. Overall, these data indicate a novel molecular and functional target for vitamin C impacting on glutamate signaling in retinal neurons.


Assuntos
Ácido Ascórbico/farmacologia , Glutamatos/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Retina/efeitos dos fármacos , Retina/metabolismo , Vitaminas/farmacologia , Animais , Biotinilação , Células Cultivadas , Embrião de Galinha , Galinhas , Transportador 3 de Aminoácido Excitatório/metabolismo , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA