Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Chin J Traumatol ; 27(1): 18-26, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37423838

RESUMO

PURPOSE: The incidence of heatstroke (HS) is not particularly high; however, once it occurs, the consequences are serious. It is reported that calcitonin gene-related peptide (CGRP) is protective against brain injury in HS rats, but detailed molecular mechanisms need to be further investigated. In this study, we further explored whether CGRP inhibited neuronal apoptosis in HS rats via protein kinase A (PKA)/p-cAMP response element-binding protein (p-CREB) pathway. METHODS: We established a HS rat model in a pre-warmed artificial climate chamber with a temperature of (35.5 ± 0.5) °C and a relative humidity of 60% ± 5%. Heatstress was stopped once core body temperature reaches above 41 °C. A total of 25 rats were randomly divided into 5 groups with 5 animals each: control group, HS group, HS+CGRP group, HS+CGRP antagonist (CGRP8-37) group, and HS+CGRP+PKA/p-CREB pathway blocker (H89) group. A bolus injection of CGRP was administered to each rat in HS+CGRP group, CGRP8-37 (antagonist of CGRP) in HS+CGRP8-37 group, and CGRP with H89 in HS+CGRP+H89 group. Electroencephalograms were recorded and the serum concentration of S100B, neuron-specific enolase (NSE), neuron apoptosis, activated caspase-3 and CGRP expression, as well as pathological morphology of brain tissue were detected at 2 h, 6 h, and 24 h after HS in vivo. The expression of PKA, p-CREB, and Bcl-2 in rat neurons were also detected at 2 h after HS in vitro. Exogenous CGRP, CGRP8-37, or H89 were used to determine whether CGRP plays a protective role in brain injury via PKA/p-CREB pathway. The unpaired t-test was used between the 2 samples, and the mean ± SD was used for multiple samples. Double-tailed p < 0.05 was considered statistically significant. RESULTS: Electroencephalogram showed significant alteration of θ (54.50 ± 11.51 vs. 31.30 ± 8.71, F = 6.790, p = 0.005) and α wave (16.60 ± 3.21 vs. 35.40 ± 11.28, F = 4.549, p = 0.020) in HS group compared to the control group 2 h after HS. The results of triphosphate gap terminal labeling (TUNEL) showed that the neuronal apoptosis of HS rats was increased in the cortex (9.67 ± 3.16 vs. 1.80 ± 1.10, F = 11.002, p = 0.001) and hippocampus (15.73 ± 8.92 vs. 2.00 ± 1.00, F = 4.089, p = 0.028), the expression of activated caspase-3 was increased in the cortex (61.76 ± 25.13 vs. 19.57 ± 17.88, F = 5.695, p = 0.009) and hippocampus (58.60 ± 23.30 vs. 17.80 ± 17.62, F = 4.628, p = 0.019); meanwhile the expression of serum NSE (5.77 ± 1.78 vs. 2.35 ± 0.56, F = 5.174, p = 0.013) and S100B (2.86 ± 0.69 vs. 1.35 ± 0.34, F = 10.982, p = 0.001) were increased significantly under HS. Exogenous CGRP decreased the concentrations of NSE and S100B, and activated the expression of caspase-3 (0.41 ± 0.09 vs. 0.23 ± 0.04, F = 32.387, p < 0.001) under HS; while CGRP8-37 increased NSE (3.99 ± 0.47 vs. 2.40 ± 0.50, F = 11.991, p = 0.000) and S100B (2.19 ± 0.43 vs. 1.42 ± 0.30, F = 4.078, p = 0.025), and activated the expression caspase-3 (0.79 ± 0.10 vs. 0.23 ± 0.04, F = 32.387, p < 0.001). For the cell experiment, CGRP increased Bcl-2 (2.01 ± 0.73 vs. 2.15 ± 0.74, F = 8.993, p < 0.001), PKA (0.88 ± 0.08 vs. 0.37 ± 0.14, F = 20.370, p < 0.001), and p-CREB (0.87 ± 0.13 vs. 0.29 ± 0.10, F = 16.759, p < 0.001) levels; while H89, a blocker of the PKA/p-CREB pathway reversed the expression. CONCLUSIONS: CGRP can protect against HS-induced neuron apoptosis via PKA/p-CREB pathway and reduce activation of caspase-3 by regulating Bcl-2. Thus CGRP may be a new target for the treatment of brain injury in HS.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina , Golpe de Calor , Isoquinolinas , Sulfonamidas , Animais , Ratos , Apoptose , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Caspase 3 , Proteínas Proto-Oncogênicas c-bcl-2 , Ratos Sprague-Dawley , Golpe de Calor/metabolismo , Golpe de Calor/patologia
2.
Int Immunopharmacol ; 126: 111305, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-38043264

RESUMO

BACKGROUND: Severe heat stroke is often complicated by multiple organ failure, including liver injury. Recent evidence indicates that the underlying mechanism constitutes sterile inflammation triggered by cell damage, in which hepatocyte NOD-like receptor family pyrin domain-containing 3 inflammasome activation and pyroptosis play key roles. As extracellular histones act as damage-associated molecular patterns and mediate tissue toxicity and inflammation, we aimed to investigate whether extracellular histones contribute to inducing hepatocyte pyroptosis following heat stroke, promoting the development of liver inflammation and injury, and elucidate the potential underlying mechanisms. METHODS: Exogenous histones were administered to AML-12 murine hepatocytes or male aged 8-12 week mice following hyperthermic treatment (at 39 °C in a chamber with 60 % relative humidity). Prior to heat exposure, endogenous histones were neutralized using neutralizing antibodies, inflammasomes were inhibited by RNA silencing, and Toll-like receptor 9 was modulated using a pharmacological agonist or antagonist. Inflammasome assembly, caspase-1 activation, histological changes, and liver enzyme levels were measured. Statistical comparison of more than two groups was performed using one-way ANOVA with Tukey's post-hoc testing. The correlations were analyzed using Pearson's correlation test. All experiments were repeated thrice. A p-value < 0.05 was considered significant. RESULTS: Heat stroke induced histone release into the extracellular space at levels correlating with liver injury. Moreover, extracellular histones augmented heat stroke-induced liver injury both in vitro and in vivo in a dose- and time-dependent manner, whereas neutralizing histones conferred protection following heat stroke. Histones mediated NOD-like receptor family pyrin domain-containing 3 inflammasome activation through the Toll-like receptor 9 signaling pathway, which resulted in hepatocyte pyroptosis and liver inflammation. CONCLUSIONS: Our findings show that histones are critical mediators of hepatocyte pyroptosis that aggravate liver injury in a heat stroke setting. Therefore, we suggest extracellular histones as potential therapeutic targets to limit heat stroke-induced cell death and liver injury.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Golpe de Calor , Hepatite , Masculino , Camundongos , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Histonas/metabolismo , Inflamassomos/metabolismo , Piroptose , Receptor Toll-Like 9/metabolismo , Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Hepatócitos/metabolismo , Inflamação , Hepatite/patologia , Golpe de Calor/complicações , Golpe de Calor/patologia
3.
Curr Stem Cell Res Ther ; 18(3): 401-409, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35546753

RESUMO

BACKGROUND: Heatstroke (HS) is a serious disease caused by central nervous system (CNS) injuries, such as delirium, convulsion, and coma. Currently, mesenchymal stem cells (MSCs) have demonstrated novel neuroprotective effects; therefore, this research explores the neuroprotective effects and mechanisms of MSCs against HS injury. METHODS: HS rat models were induced in a 40°C and 65% humidity environment until the rectal temperature reached 42°C. The verified HS injury model rats were divided into the HS and MSCs-treated groups. Each rat in the treated group was infused with 1x106 MSCs suspended in 0.3 ml physiological saline via the tail vein. The HS- or MSCs-treated rats were further divided into early-stage (3d) and late-stage (28d). HS rat models were induced by a high-temperature and high-humidity environment at a specific time, the mortality was analyzed, and an automatic biochemical analyzer measured levels of liver and kidney function indicators in the blood. The neurons' morphologic changes were observed through Nissl staining, and neurological deficit scores were performed. Moreover, the levels of inflammatory factors in brain tissue were measured using a multi-cytokine detection platform, and the expression of BDNF, phosphorylated TrkB and P38 were detected by the Western Bolt. RESULTS: MSCs injection significantly reduced mortality and alleviated liver and kidney function. Moreover, the neurological deficit and neuronic edema of the hippocampus caused by HS at 3d and 28d were significantly ameliorated by MSCs administration. Specifically, the injection of MSCs inhibited high levels of interleukin (IL)-1ß, IL-6, tumor necrosis factor-α (TNF-α), and IL-17A caused by HS but elevated the levels of IL-10 and IL-13 in the early period (3d); while in the later period (28d), MSCs significantly increased the levels of IL-10 and IL-13 continuously and inhibited the high level of IL-17A. Furthermore, MSCs injection increased the expressions of BDNF and phosphorylated TrkB (BDNF receptor), meanwhile inhibiting the expression of phosphorylated P38 (inflammatory factor) in the brains of HS rats in the early period (3d) but had no significant influence on the later period (28d). CONCLUSION: These results suggested that MSCs injection may provide therapeutic effects for HS in rats by improving liver and kidney function and reducing CNS damage. Moreover, MSCs injection inhibited the brain inflammatory response of HS rats, and the BDNF-TrkB and P38/MAPK signal pathways may be involved, providing a potential mechanism for HS therapy by MSCs administration.


Assuntos
Golpe de Calor , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Fármacos Neuroprotetores , Ratos , Animais , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Interleucina-13/metabolismo , Encéfalo , Golpe de Calor/terapia , Golpe de Calor/metabolismo , Golpe de Calor/patologia , Células-Tronco Mesenquimais/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos
4.
J Cachexia Sarcopenia Muscle ; 13(3): 1717-1730, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35243801

RESUMO

BACKGROUND: Rhabdomyolysis (RM) is a common complication of exertional heat stroke (EHS) and constitutes a direct cause of death. However, the mechanism underlying RM following EHS remains unclear. METHODS: The murine EHS model was prepared by our previous protocol. RNA sequencing is applied to identify the pathological pathways that contribute to RM following EHS. Inhibition of the acyl-CoA synthetase long-chain family member 4 (ACSL4) was achieved by RNA silencing in vitro prior to ionomycin plus heat stress exposure or pharmacological inhibitors in vivo prior to heat and exertion exposure. The histological changes, the iron accumulation, oxidized phosphatidylethanolamines species, as well as histological evaluation and levels of lipid metabolites in skeletal muscle tissues were measured. RESULTS: We demonstrated that ferroptosis contributes to RM development following EHS. Ferroptosis inhibitor ferrostatin-1 administration once EHS onset significantly ameliorated the survival rate of EHS mice from 35.357% to 52.288% within 24 h after EHS (P = 0.0028 compared with control) and markedly inhibited RM development induced by EHS. By comparing gene expression of between sham heat rest (SHR) (n = 3) and EHS (n = 3) mice in the gastrocnemius (Gas) muscle tissue, we identified that Acsl4 mRNA expression is elevated in Gas muscle tissue of EHS mice (P = 0.0038 compared with SHR), so as to its protein levels (P = 0.0001 compared with SHR). Followed by increase in creatine kinase (CK) and myoglobin (MB) levels, the labile iron accumulation, decrease in glutathione peroxidase 4 (GPX4) expression, and elevation of lipid peroxidation products. From in vivo and in vitro experiments, inhibition of Acsl4 significantly improves muscle cell death caused by EHS, thereby ameliorating RM development, followed by reduction in CK and MB levels by 30-40% (P < 0.0001; n = 8-10) and 40% (P < 0.0001; n = 8-10), restoration of GPX4 expression, and decrease in lipid peroxidation products. Mechanistically, ACSL4-mediated RM seems to be Yes-associated protein (YAP) dependent via TEA domain transcription factor1/TEA domain transcription factor4. CONCLUSIONS: These findings demonstrate an important role of ACSL4 in mediating ferroptosis activation in the development of RM following EHS and suggest that targeting ACSL4 may represent a novel therapeutic strategy to limit the skeletal muscle cell death and prevent RM after EHS.


Assuntos
Coenzima A Ligases , Ferroptose , Golpe de Calor , Rabdomiólise , Animais , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Golpe de Calor/genética , Golpe de Calor/metabolismo , Golpe de Calor/patologia , Ferro/metabolismo , Camundongos , Rabdomiólise/genética , Rabdomiólise/metabolismo , Rabdomiólise/patologia
5.
J Therm Biol ; 101: 103081, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34879909

RESUMO

Heat stroke is a severe systemic inflammatory response disease caused by high fever, mainly with nervous system damage. Mesenchymal stem cells (MSCs) are currently believed to have anti-inflammation and immunomodulatory effects. Therefore, we aimed to explore the protective effect and mechanism of MSCs on heat stroke-induced excessive inflammation and neurological dysfunction. We established a heat stroke model in rats under conditions of continuous high temperature and high humidity. After modeling, rats were randomly divided into heat stroke model group, MSCs treatment group and normal temperature control group without any treatment. We performed survival analysis, neurological deficit score, histological staining of hippocampus and cerebellum, immunofluorescence staining of microglia, detection of inflammatory and chemokine levels in the hippocampus and cerebellum in each group. We found that MSCs treatment not only significantly reduced early (day 3) and late (day 28) mortality, but also prominently reduced nerve injury in heat stroke rats, and improved pathology and neuronal cell damage in the hippocampus and cerebellum. In addition, MSCs treatment can significantly inhibit the over-activation of hippocampal microglia in heat stroke rats and the levels of pro-inflammatory factors and chemokines in the hippocampus. Early treatment of MSCs can greatly promote the activation of cerebellar microglia in heat stroke rats. Meanwhile, MSCs treatment has an inhibitory effect on the level of chemokine in the cerebellum of rats in the early stage of heat stroke. In conclusion, the application of MSCs in the treatment of heat stroke in rats can significantly reduce mortality and neurological deficits and improve hippocampal damage, possibly by inhibiting the excessive activation of hippocampal microglia in heat stroke rats.


Assuntos
Golpe de Calor/terapia , Hipocampo/patologia , Transplante de Células-Tronco Mesenquimais , Microglia , Animais , Cerebelo/imunologia , Cerebelo/patologia , Citocinas/imunologia , Golpe de Calor/imunologia , Golpe de Calor/patologia , Hipocampo/imunologia , Masculino , Células-Tronco Mesenquimais , Ratos Sprague-Dawley
6.
Shock ; 56(4): 593-600, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34524269

RESUMO

ABSTRACT: Heat stroke is characterized by excessive oxidative stress and inflammatory responses, both of which are implicated in vascular endothelial glycocalyx shedding and heat-stroke mortality. Although molecular hydrogen has antioxidation and anti-inflammatory potency, its effect on the vascular endothelial glycocalyx in heat stroke has not been examined. Therefore, the aim of this study was to investigate the influence of hydrogen inhalation on the survival and thickness of the vascular endothelial glycocalyx of rats subjected to heat stroke. Altogether, 98 Wistar rats were assigned to the experiments. A heat-controlled chamber set at 40°C temperature and 60% humidity was used to induce heat stroke. After preparation, the anesthetized rats that underwent the heating process were subjected to an hour of stabilization in which 0%, 2%, or 4% hydrogen gas was inhaled and maintained until the experiment ended. In addition to survival rate assessments, blood samples and left ventricles were collected to evaluate the thickness of the vascular endothelial glycocalyx and relevant biomarkers. The results showed that 2% hydrogen gas significantly improved survival in the heat-stroked rats and partially preserved the thickness of the endothelial glycocalyx. In addition, serum levels of endotoxin, syndecan-1, malondialdehyde, and tumor necrosis factor-α decreased, whereas superoxide dismutase levels increased, indicating that inhalation of 2% hydrogen attenuated the damage to the vascular endothelial glycocalyx through its antioxidative and anti-inflammatory effects.


Assuntos
Deutério/administração & dosagem , Células Endoteliais/efeitos dos fármacos , Glicocálix/efeitos dos fármacos , Golpe de Calor/metabolismo , Golpe de Calor/terapia , Administração por Inalação , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Glicocálix/metabolismo , Golpe de Calor/patologia , Masculino , Ratos , Ratos Wistar
7.
Int J Hyperthermia ; 37(1): 517-530, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32423248

RESUMO

Background: Worldwide heat stroke incidence has increased in recent years and is associated with high morbidity and mortality. Therefore, it is critical to identify mechanisms that mediate heat stroke. Previous studies suggested that damage to the small intestine may be a major factor in heat stroke-related morbidity and mortality. However, the mechanism underlying heat stroke related small intestine injury remains unclear.Methods: To explore how heat stroke promotes intestinal damage, we applied two well established models: mouse and IEC-6 cells heat stress (HS) to mimic heat stroke both in vivo and in vitro. The percentages of viability and cell death were assessed by WST-1 and LDH release assays. Induction of HS-induced cell death was analyzed by flow cytometry with Annexin V-FITC/PI staining. Flow cytometry was used to analyze HS-induced mitochondrial superoxide with MitoSOX staining. Malondialdehyde (MDA) levels and superoxide dismutase (SOD) levels were detected by ELISA. Flow cytometry was used to analyze HS-induced mitochondrial depolarization (low ΔΨm) with JC-1 staining. Histopathology changes in the ileum were detected by H&E staining.The ileum ultrastructure was observed by transmission electron microscopy (TEM). RIPK1, RIPK3, phosphorylated MLKL, and MLKL levels were detected by Western blot. RIPK1-RIPK3 complexes were measured by immunoprecipitation assay.Results: HS increased both necrotic cell rate and RIPK1, RIPK3, and phosphorylated MLKL expression levels in IEC-6 cells. These increased expression levels promoted higher RIPK1-RIPK3 complex formation, leading to necrosome formation both in vivo and in vitro. Moreover, HS caused dyshomeostasis, an oxidative stress response, and mitochondrial damage, along with small intestinal tissue injury and cell death. However, IEC-6 cells or mice pretreated with the RIPK1 activity chemical inhibitor Nec-1 or RIPK3 activity chemical inhibitor GSK'872 significantly reversed these phenomena and promoted balance in oxidative stress response homeostasis. More importantly, the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) pretreatment significantly inhibited HS-induced RIPK1/RIPK3-dependent necroptosis formation both in vivo and in vitro, suggesting that preventing necroptosis via scavenging ROS production might alleviate HS-induced small intestinal tissue injury and cell death.Conclusion: This study provides strong evidence that HS causes damage to both the small intestine and intestinal epithelial cells, scavenging ROS production can significantly alleviate such RIPK1/RIPK3-dependent necroptosis, mediating HS-induced intestinal damage both in vitro and in vivo. These findings provide a clear target for future mechanism-based therapeutic strategies for patients diagnosed with heat stroke.


Assuntos
Golpe de Calor/complicações , Resposta ao Choque Térmico/imunologia , Intestinos/patologia , Necroptose/imunologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Golpe de Calor/patologia , Humanos , Camundongos
8.
Mediators Inflamm ; 2019: 8187529, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31885498

RESUMO

Heat stroke (HS) is a life-threatening illness and defined as when body temperature elevates above 40°C accompanied by the systemic inflammatory response syndrome that results in multiple organ dysfunctions. α-Lipoic acid (ALA) acts as a cofactor of mitochondrial enzymes and exerts anti-inflammatory and antioxidant properties in a variety of diseases. This study investigates the beneficial effects of ALA on myocardial injury and organ damage caused by experimental HS and further explores its underlying mechanism. Male Wistar rats were exposed to 42°C until their rectal core temperature reached 42.9°C and ALA was pretreared 40 or 80 mg/kg (i.v.) 1.5 h prior to heat exposure. Results showed that HS-induced lethality and hypothermia were significantly alleviated by ALA treatment that also improved plasma levels of CRE, LDH, and CPK and myocardial injury biomarkers myoglobin and troponin. In addition, ALA reduced cardiac superoxide anion formation and protein expression of cleaved caspase 3 caused by HS. Proinflammatory cytokine TNF-α and NF-κB pathways were significantly reduced by ALA treatment which may be associated with the upregulation of Hsp70. ALA significantly increased the Atg5-12 complex and LC3B II/LC3B I ratio, whereas the p62 and p-mTOR expression was attenuated in HS rats, indicating the activation of autophagy by ALA. In conclusion, ALA ameliorated the deleterious effects of HS by exerting antioxidative and anti-inflammatory capacities. Induction of Hsp70 and activation of autophagy contribute to the protective effects of ALA in HS-induced myocardial injury.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Golpe de Calor/tratamento farmacológico , Golpe de Calor/patologia , Inflamação/tratamento farmacológico , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Ácido Tióctico/uso terapêutico , Animais , Autofagia/fisiologia , Proteínas de Choque Térmico HSP70/metabolismo , Golpe de Calor/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Masculino , NF-kappa B/metabolismo , Ratos , Ratos Wistar , Superóxidos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
9.
Biochem Biophys Res Commun ; 506(3): 626-631, 2018 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-30454698

RESUMO

Heat stroke is a serious pathological condition with high mortality. Vascular endothelial cell injury is a key feature in the pathogenesis of heat stroke, but the specific pathophysiological process whereby this occurs is still unclear. Currently, relevant studies are primarily based upon examination of apoptosis. Recently, pyroptosis, a new form of inflammation-related programmed cell death, was also demonstrated to be involved in heat stroke pathophysiology. Herein, we present evidence that vascular endothelial cell pyroptosis can be induced by heat stress in a time- and temperature-dependent manner. Furthermore, this process can be significantly inhibited by GSDMD siRNA. These findings suggest a new therapeutic target for heat stroke.


Assuntos
Golpe de Calor/patologia , Células Endoteliais da Veia Umbilical Humana/patologia , Piroptose , Caspase 1/metabolismo , Golpe de Calor/metabolismo , Resposta ao Choque Térmico , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Neoplasias/metabolismo , Proteínas de Ligação a Fosfato , RNA Interferente Pequeno/metabolismo
10.
Cell Mol Biol (Noisy-le-grand) ; 64(7): 36-42, 2018 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-29974844

RESUMO

Heatstroke is still a potentially fatal threat during summer heat waves, despite improved prevention and treatment. It is reported that the transient receptor potential vanilloid 4 (TRPV4) inhibitor may protect septicemia mice. Many aspects of heatstroke have been defined, from the sepsis-mimic inflammatory response to hyperthermia. Hence, TRPV4 may be a therapeutic target for heatstroke. The results in murine models of heatstroke verified that GSK2193874, as a selected TRPV4 inhibitor, was injected at heatstroke onset, and then reduced the reduction of core temperature, the death rate, wet/dry ratio of the lung, levels of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6, coagulation indicators, the degree of organ injury, and caspase-3/7 activity (P<0.05). But GSK2193874 treatment before heat stress did not improve the symptoms of heatstroke mice. Therefore, TRPV4 should be involved in heatstroke-induced injury. Timely GSK2193874 administration may be useful to reduce heatstroke-induced injury. TRPV4 may be a potential new therapeutic target in fatal heatstroke.


Assuntos
Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Golpe de Calor/tratamento farmacológico , Piperidinas/farmacologia , Edema Pulmonar/tratamento farmacológico , Quinolinas/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Animais , Anti-Inflamatórios/uso terapêutico , Caspase 3/metabolismo , Caspase 7/metabolismo , Modelos Animais de Doenças , Golpe de Calor/complicações , Golpe de Calor/patologia , Temperatura Alta/efeitos adversos , Interleucina-6/sangue , Interleucina-6/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piperidinas/uso terapêutico , Edema Pulmonar/etiologia , Edema Pulmonar/patologia , Quinolinas/uso terapêutico , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia
11.
Mol Med Rep ; 16(1): 87-94, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28498471

RESUMO

The aim of the present study was to investigate the effects of sodium tanshinone IIA sulfonate (STS) on inflammatory responses, aortic endothelial cell apoptosis, disseminated intravascular coagulation (DIC) and multiple organ damage in an animal model of classic heat stroke (CHS). The rats in the heat stroke (HS) and STS­treated heat stroke (STS­HS) groups were placed into a pre­warmed animal temperature controller (ATC) at 35˚C. The moment at which the rectal temperature reached 43.5˚C was considered as the time of onset of HS. In the HS groups, the rats were removed from the ATC and allowed to recover at 26˚C for 0, 2, 6 or 12 h. In the STS­HS groups, the rats received femoral vein injections of 5­40 mg/kg STS immediately following the onset of HS and were subsequently placed at a temperature of 26˚C to recover for 6 h. In the present study, the serum levels of tumor necrosis factor (TNF)­α, interleukin (IL)­1ß and IL­6 were assessed using ELISA, and the numbers of apoptotic aortic endothelial cells were investigated using terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick­end labeling combined with immunofluorescence. In the HS groups, the serum levels of TNF­α, IL­1ß and IL­6, as well as the numbers of apoptotic aortic endothelial cells were increased compared with the normothermic control group. Additionally, the plasma prothrombin time, activated partial thromboplastin time and D­dimer level were significantly increased in the HS group compared with the normothermic control group following recovery for 6 h. By contrast, the platelet count was decreased in the HS group compared with the normothermic control group. The serum levels of creatinine, blood urea nitrogen, alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase and lactate dehydrogenase were increased and histopathological damage to multiple organs was observed in the HS group following recovery for 6 h. In the STS­HS groups, cytokine levels and apoptotic aortic endothelial cell numbers were reduced compared with the HS group after 6 h recovery. STS (40 mg/kg) treatment additionally improved the serum levels of organ injury indicators and plasma indicators of coagulopathy, and prevented histopathological damage to multiple organs. These findings demonstrated that STS treatment may ameliorate multiple organ damage by attenuating inflammatory responses, aortic endothelial cell apoptosis and DIC in CHS. These results suggested that STS may hold potential as an alternative therapeutic strategy for the treatment of patients with HS.


Assuntos
Apoptose/efeitos dos fármacos , Coagulação Intravascular Disseminada/sangue , Coagulação Intravascular Disseminada/etiologia , Células Endoteliais/efeitos dos fármacos , Golpe de Calor/complicações , Golpe de Calor/patologia , Fenantrenos/farmacologia , Animais , Biomarcadores , Citocinas/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Coagulação Intravascular Disseminada/tratamento farmacológico , Golpe de Calor/tratamento farmacológico , Inflamação/patologia , Masculino , Ratos
12.
Int J Clin Exp Pathol ; 8(6): 6732-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26261556

RESUMO

Heatstroke not only directly induces cell injury, but also causes large amounts of inflammatory mediators release and cells with extensive biological activities to induce a systemic inflammatory response and immune dysfunction. This study aimed to observe the effects of JAK2 inhibitor AG490 on the brain injury and inflammatory responses of rats with systemic heatstroke. Under the light microscope, the hippocampus tissues of rat with heatstroke were edema and apoptotic rate was increased. Up-regulation of malondialdehyde (MDA), nitric oxide synthase (iNOS), reactive oxygen species (ROS) and down-regulation of superoxide dismutase (SOD) were also found after heatstroke in rats, which compared with that of the control group. Heatstroke induced inflammation factors secretions and up-regulated levels of matrix metallopeptidase 2 and 9 (MMP2 and MMP-9) and systemic inflammatory response molecules including intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-beta 1 (TNF-ß1) and cyclooxygenase-2 (COX-2). However, the JAK2 inhibitor AG490 was significantly attenuated the brain injury and inflammatory responses induced by heatstroke in rats. The survival time of heatstroke rats showed that AG490 notably lived longer than heatstroke rats without AG490 treatment. These findings suggest that AG490 may prevent the occurrence of heatstroke via inhibiting the JAK2/STAT3 pathway and the systemic inflammatory responses.


Assuntos
Anti-Inflamatórios/farmacologia , Golpe de Calor/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Inflamação/prevenção & controle , Janus Quinase 2/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT3/metabolismo , Tirfostinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Edema Encefálico/enzimologia , Edema Encefálico/imunologia , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Ciclo-Oxigenase 2/metabolismo , Golpe de Calor/enzimologia , Golpe de Calor/imunologia , Golpe de Calor/patologia , Hipocampo/enzimologia , Hipocampo/imunologia , Hipocampo/patologia , Inflamação/enzimologia , Inflamação/imunologia , Inflamação/patologia , Molécula 1 de Adesão Intercelular/metabolismo , Janus Quinase 2/metabolismo , Masculino , Malondialdeído/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo
13.
BMC Complement Altern Med ; 15: 4, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25653103

RESUMO

BACKGROUND: The pathogenesis of heatstroke is a multi-factorial process involved with an interplay among subsequent inflammation, endothelial injury and coagulation disturbances, which makes pharmacological therapy of heatstroke a challenging problem. Xuebijing injection (XBJ), a traditional Chinese medicine used to sepsis, has been reported to suppress inflammatory responses and restore coagulation disturbances. However, little is known about the role of XBJ in heatstroke. METHODS: Mice were treated with indicated dose of XBJ before and/or after the induction of heatstroke. Serum inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), and endothelial markers, von Willebrand Factor (vWF) and E-selectin, were measured by ELISA. Liver, kidney and heart profiles including alanine aminotransferase, aspartic aminotransferase, creatinine, blood urea nitrogen, and lactate dehydrogenase, were evaluated by UniCel DxC 800 Synchron Clinical Systems, and troponin was measured by ELISA. Coagulation profiles, including thrombin time, prothrombin time, activated partial thromboplastin time, international normalized ratio, and fibrinogen were examined by STA Compact® Hemostasis System. Jejunum injury was evaluated with H&E staining. Changes in mitochondrial structure in cardiac tissue were assesed by electron microscopy. RESULTS: Pretreatment with XBJ decreased serum pro-inflammatory cytokines including TNF-α and IL-6, as well as endothelial injury markers, vWF and E-selectin, in a dose-dependent manner in heatstroke mice. Similar protective effects were observed when XBJ was administered after, or both before and after heat insult. These protective effects lasted for over 12 h in mice receiving XBJ before and after heat insult. XBJ also improved survival rates in heatstroke mice, ameliorated liver, heart, and kidney injuries, including mitochondrial damage to the heart, and reduced coagulation disturbances. CONCLUSIONS: XBJ prevents organ injuries and improves survival in heatstroke mice by attenuating inflammatory responses and endothelial injury. XBJ may be a potentially useful in the prevention and treatment of heatstroke.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Endotélio Vascular/efeitos dos fármacos , Coração/efeitos dos fármacos , Golpe de Calor/tratamento farmacológico , Inflamação/prevenção & controle , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Animais , Coagulação Sanguínea , Citocinas/sangue , Medicamentos de Ervas Chinesas/farmacologia , Selectina E/sangue , Endotélio Vascular/patologia , Ensaio de Imunoadsorção Enzimática , Golpe de Calor/sangue , Golpe de Calor/mortalidade , Golpe de Calor/patologia , Inflamação/sangue , Inflamação/etiologia , Interleucina-6/sangue , Masculino , Medicina Tradicional Chinesa , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/patologia , Fitoterapia , Fator de Necrose Tumoral alfa/sangue , Fator de von Willebrand/metabolismo
14.
CNS Neurol Disord Drug Targets ; 14(3): 386-99, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25714976

RESUMO

We have shown previously that heat stroke produced by whole body hyperthermia (WBH) for 4 h at 38°C in diabetic rats exacerbates blood-brain barrier breakdown, brain edema formation and neuronal cell injury as compared to healthy animals after identical heat exposure. In this combination of diabetes and WBH, normal therapeutic measures do not induce sufficient neuroprotection. Thus, we investigated whether nanowired mesenchymal cells (MSCs) when delivered systemically may have better therapeutic effects on brain damage in diabetic rats after WBH. Diabetes induced by streptozotocin administration (75 mg/kg, i.p, daily for 3 days) in rats resulted in clinical symptoms of the disease within 4 to 6 weeks (blood glucose level 20 to 30 mmoles/l as compared to saline control groups (4 to 6 mmoles/l). When subjected to WBH, these diabetic rats showed a 4-to 6-fold exacerbation of blood-brain barrier breakdown to Evans blue and radioiodine, along with brain edema formation and neuronal cell injury. Intravenous administration of rat MSCs (1x10(6)) to diabetic rats one week before WBH slightly reduced brain pathology, whereas TiO2 nanowired MSCs administered in an identical manner resulted in almost complete neuroprotection. On the other hand, MSCs alone significantly reduced brain pathology in saline-treated rats after WBH. These observations indicate that nanowired delivery of stem cells has superior therapeutic potential in heat stroke with diabetes, pointing to novel clinical perspectives in the future.


Assuntos
Encéfalo/patologia , Diabetes Mellitus Experimental/patologia , Golpe de Calor/patologia , Golpe de Calor/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Axônios/fisiologia , Axônios/ultraestrutura , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/fisiopatologia , Edema Encefálico/patologia , Edema Encefálico/fisiopatologia , Permeabilidade Capilar/fisiologia , Diabetes Mellitus Experimental/fisiopatologia , Marcha/fisiologia , Golpe de Calor/fisiopatologia , Masculino , Transplante de Células-Tronco Mesenquimais/instrumentação , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/fisiologia , Nanopartículas Metálicas , Bainha de Mielina/fisiologia , Bainha de Mielina/ultraestrutura , Ratos Sprague-Dawley , Titânio , Caminhada/fisiologia
15.
PLoS One ; 8(2): e56728, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23424673

RESUMO

This study was performed to gain insights into novel therapeutic approaches for the treatment of heatstroke. The central nervous system regulates peripheral immune responses via the vagus nerve, the primary neural component of the cholinergic anti-inflammatory pathway. Electrical vagus nerve stimulation (VNS) reportedly suppresses pro-inflammatory cytokine release in several models of inflammatory disease. Here, we evaluated whether electrical VNS attenuates severe heatstroke, which induces a systemic inflammatory response. Anesthetized rats were subjected to heat stress (41.5°C for 30 minutes) with/without electrical VNS. In the VNS-treated group, the cervical vagus nerve was stimulated with constant voltage (10 V, 2 ms, 5 Hz) for 20 minutes immediately after completion of heat stress. Sham-operated animals underwent the same procedure without stimulation under a normothermic condition. Seven-day mortality improved significantly in the VNS-treated group versus control group. Electrical VNS significantly suppressed induction of pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-6 in the serum 6 hours after heat stress. Simultaneously, the increase of soluble thrombomodulin and E-selectin following heat stress was also suppressed by VNS treatment, suggesting its protective effect on endothelium. Immunohistochemical analysis using tissue preparations obtained 6 hours after heat stress revealed that VNS treatment attenuated infiltration of inflammatory (CD11b-positive) cells in lung and spleen. Interestingly, most cells with increased CD11b positivity in response to heat stress did not express α7 nicotinic acetylcholine receptor in the spleen. These data indicate that electrical VNS modulated cholinergic anti-inflammatory pathway abnormalities induced by heat stress, and this protective effect was associated with improved mortality. These findings may provide a novel therapeutic strategy to combat severe heatstroke in the critical care setting.


Assuntos
Terapia por Estimulação Elétrica/métodos , Golpe de Calor/complicações , Golpe de Calor/terapia , Nervo Vago , Animais , Antígeno CD11b/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Golpe de Calor/metabolismo , Golpe de Calor/patologia , Resposta ao Choque Térmico , Inflamação/complicações , Pulmão/patologia , Masculino , Ratos , Ratos Wistar , Receptores Nicotínicos/metabolismo , Baço/patologia , Análise de Sobrevida , Receptor Nicotínico de Acetilcolina alfa7
16.
Eur J Pharmacol ; 688(1-3): 62-7, 2012 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-22609231

RESUMO

Flutamide has been used as an adjunct for decreasing the mortality from subsequent sepsis. Heatstroke resembles septic shock in many aspects. We hypothesized that heat-induced multiple organ dysfunction syndromes and lethality could be reduced by flutamide therapy. In heatstroke groups, mice were exposed to whole body heating (41.2°C, for 1h) in a controlled-environment chamber. The heat-stressed mice were returned to normal room temperature (24°C) after whole body heating. Mice still alive on day 4 of WBH treatment were considered survivors. Physiological and biochemical parameters were monitored for 2.5h post-WBH. Heatstroke mice were subcutaneously treated with flutamide (12.5-50mg/kg body weight in 0.05 ml) or vehicle solution (0.05 ml/kg body weight) once daily for 3 consecutive days post-WBH. We evaluated the effect of flutamide in heatstroke mice and showed that flutamide significantly (i) attenuated hypothermia, (ii) reduced the number of apoptotic cells in the hypothalamus, the spleen, the liver, and the kidney, (iii) attenuated the plasma index of toxic oxidizing radicals (e.g., nitric oxide metabolites and hydroxyl radicals), (iv) diminished the plasma index of the organ injury index (e.g., lactate dehydrogenase), (v) attenuated plasma systemic inflammation response molecules (e.g., tumor necrosis factor-α and interleukin-6), (vi) reduced the index of infiltration of polymorphonuclear neutrophils in the lung (e.g., myeloperoxidase activity), and (vii) allowed three times the fractional survival compared with vehicle. Thus, flutamide appears to be a novel agent for the treatment of mice with heatstroke or patients in the early stage of heatstroke.


Assuntos
Antagonistas de Receptores de Andrógenos/farmacologia , Flutamida/farmacologia , Golpe de Calor/tratamento farmacológico , Receptores Androgênicos/metabolismo , Antagonistas de Receptores de Andrógenos/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Regulação da Temperatura Corporal/efeitos dos fármacos , Castração , Citocinas/sangue , Flutamida/uso terapêutico , Golpe de Calor/sangue , Golpe de Calor/patologia , Golpe de Calor/fisiopatologia , Hidroxibenzoatos/sangue , Hipotálamo/efeitos dos fármacos , Hipotálamo/patologia , Hipotálamo/fisiopatologia , L-Lactato Desidrogenase/sangue , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Masculino , Camundongos , Neutrófilos/efeitos dos fármacos , Óxido Nítrico/sangue , Óxido Nítrico/metabolismo , Taxa de Sobrevida
17.
Acta Pharmacol Sin ; 33(6): 775-82, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22609835

RESUMO

AIM: To assess the therapeutic effect of melatonin on heat-induced acute lung inflammation and injury in rats. METHODS: Heatstroke was induced by exposing anesthetized rats to heat stress (36 °C, 100 min). Rats were treated with vehicle or melatonin (0.2, 1, 5 mg/kg) by intravenous administration 100 min after the initiatioin of heatstroke and were allowed to recover at room temperature (26 °C). The acute lung injury was quantified by morphological examination and by determination of the volume of pleural exudates, the number of polymorphonuclear (PMN) cells, and the myeloperoxidase (MPO) activity. The concentrations of tumor necrosis factor, interleukin (IL)-1ß, IL-6, and IL-10 in bronchoalveolar fluid (BALF) were measured by ELISA. Nitric oxide (NO) level was determined by Griess method. The levels of glutamate and lactate-to-pyruvate ratio were analyzed by CMA600 microdialysis analyzer. The concentrations of hydroxyl radicals were measured by a procedure based on the hydroxylation of sodium salicylates leading to the production of 2,3-dihydroxybenzoic acid (DHBA). RESULTS: Melatonin (1 and 5 mg/kg) significantly (i) prolonged the survival time of heartstroke rats (117 and 186 min vs 59 min); (ii) attenuated heatstroke-induced hyperthermia and hypotension; (iii) attenuated acute lung injury, including edema, neutrophil infiltration, and hemorrhage scores; (iv) down-regulated exudate volume, BALF PMN cell number, and MPO activity; (v) decreased the BALF levels of lung inflammation response cytokines like TNF-alpha, interleukin (IL)-1ß, and IL-6 but further increased the level of an anti-inflammatory cytokine IL-10; (vi) reduced BALF levels of glutamate, lactate-to-pyruvate ratio, NO, 2,3-DHBA, and lactate dehydrogenase. CONCLUSION: Melatonin may improve the outcome of heatstroke in rats by attenuating acute lung inflammation and injury.


Assuntos
Lesão Pulmonar Aguda/complicações , Lesão Pulmonar Aguda/tratamento farmacológico , Antioxidantes/uso terapêutico , Golpe de Calor/complicações , Golpe de Calor/tratamento farmacológico , Hemorragia/complicações , Hemorragia/tratamento farmacológico , Melatonina/uso terapêutico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Antioxidantes/administração & dosagem , Líquido da Lavagem Broncoalveolar/imunologia , Febre/complicações , Febre/tratamento farmacológico , Golpe de Calor/imunologia , Golpe de Calor/patologia , Hemorragia/imunologia , Hemorragia/patologia , Hipotensão/complicações , Hipotensão/tratamento farmacológico , Interleucina-10/imunologia , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Masculino , Melatonina/administração & dosagem , Óxido Nítrico/imunologia , Edema Pulmonar/complicações , Edema Pulmonar/tratamento farmacológico , Edema Pulmonar/imunologia , Edema Pulmonar/patologia , Ratos , Ratos Wistar , Resultado do Tratamento , Fator de Necrose Tumoral alfa/imunologia
18.
Am J Med Sci ; 344(5): 383-90, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22245947

RESUMO

INTRODUCTION: Heatstroke has been defined as a form of hyperthermia associated with a systemic inflammatory response that leads to multiple organ dysfunction syndrome (MODS). It has also been documented that heat shock protein 70 (HSP70) preconditioning is able to induce thermotolerance. Here, the authors further investigated whether hypobaric hypoxia preconditioning (HHP) improved the MODS in heatstroke by up-regulation of HSP70. METHODS: Anesthetized rats were randomly assigned to (a) non-HHP + nonheated group, (b) non-HHP + heated group, (c) HHP + heated group and (d) HHP + HSP70 antibodies (Abs) + heated groups. All heated groups were exposed to heat stress (43°C, 70 minutes) to induce heatstroke. For HHP, animals were exposed to 0.66 atmosphere absolute (18.3% O2) for 5 hours daily for consecutive 5 days per week for 2 weeks before the start of heat exposure. RESULTS: HHP significantly (i) attenuated hypotension, (ii) reduced plasma index of the toxic oxidizing radicals and the organ injury indicator, (iii) attenuated plasma systemic inflammatory response molecules, (iv) reduced an index of infiltration of polymorphonuclear neutrophils in the lung like myeloper-oxidase activity, (v) promoted plasma levels of an anti-inflammatory cytokine, interleukin-10, (vi) promoted the survival time to fourfold compared with non-HHP group and (vii) promoted the overexpression of HSP70 in different organs (eg, the lung) during heatstroke. The beneficial effects of HHP could be significantly attenuated by HSP70 Ab preconditioning. CONCLUSION: Our results show that HHP protects rats from heat-induced MODS via up-regulating HSP70. Thus, HHP could be a novel strategy for the prevention of heatstroke animals or patients before heat exposure.


Assuntos
Proteínas de Choque Térmico HSP70/biossíntese , Golpe de Calor/prevenção & controle , Hipóxia/patologia , Animais , Encéfalo/metabolismo , Proteínas de Choque Térmico HSP70/imunologia , Proteínas de Choque Térmico HSP70/metabolismo , Golpe de Calor/sangue , Golpe de Calor/patologia , Interleucina-10/sangue , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/sangue
19.
Eur J Pharmacol ; 661(1-3): 109-17, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21545796

RESUMO

Heatstroke is a form of excessive hyperthermia associated with a systemic inflammatory response that leads to multi-organ dysfunction in which central nervous system disorders predominate. Herein we determined to ascertain whether heat-induced multi-organ dysfunction in rats could be attenuated by granulocyte-colony stimulating factor (G-CSF) preconditioning. Anesthetized rats were divided into 2 major groups and given vehicle solution (isotonic saline, 0.3 ml, subcutaneously) or G-CSF (50-200 µg/kg body weight in 0.3 ml normal saline, subcutaneously) daily and consecutively for 5 days before the start of thermal experiments. They were exposed to an ambient temperature of 43°C for 68 min to induce heatstroke. G-CSF preconditioning significantly prolonged the survival time in heatstroke rats in a dose-related way (82-98 min vs 127-243 min). The non-preconditioning heatstroke animals showed hyperthermia, arterial hypotension, increased serum levels of systemic inflammatory response molecules, increased hypothalamic apoptotic cell numbers as well as neuronal damage scores, and increased serum levels of renal and hepatic dysfunction indicators. These heatstroke syndromes could be significantly reduced by G-CSF preconditioning. Thus our results revealed a potential for G-CSF used as a prophylactic agent for heatstroke in rats.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacologia , Golpe de Calor/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/patologia , Contagem de Células , Células Endoteliais/patologia , Febre/complicações , Febre/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Golpe de Calor/complicações , Golpe de Calor/metabolismo , Golpe de Calor/patologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Hipotensão/complicações , Hipotensão/tratamento farmacológico , Hipotálamo/patologia , Inflamação/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Fatores de Crescimento Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Análise de Sobrevida , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Biol Pharm Bull ; 33(9): 1522-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20823568

RESUMO

There is evidence that increased plasma cytokines, elevated brain levels of monoamines and hydroxyl radical production may be implicated in pathogenesis during heat stroke in rats. Acute treatment with a combined therapeutic approach has been repeatedly advocated in cerebral ischemia experiments. The aim of this study was to investigate whether the combined agent (mannitol and dexamethasone) has beneficial efficacy to improve the survival time (ST) and heat stroke-induced damage in experimental heat stroke. Urethane-anesthetized rats underwent instrumentation for the measurement of colonic temperature, mean arterial pressure (MAP), striatal cerebral blood flow (CBF), heart rate, and neuronal damage score. The rats were exposed to an ambient temperature (43 degrees C) to induce heat stroke. Concentrations of the ischemic and damage markers, dopamine, serotonin, and hydroxyl radical production in corpus striatum, and the plasma levels of tumor necrosis factor-alpha (TNF-alpha) were observed during heat stroke. After the onset of heat stroke, the heat stroke rats display decreased MAP, decreased CBF, increased the plasma levels of TNF-alpha, increased cerebral striatal monoamines and hydroxyl radical production release, and severe cerebral ischemia and neuronal damage compared with those of normothermic control rats. However, immediate treatment with the combined agent confers significant protection against heat stroke-induced arterial hypotension, systemic inflammation, cerebral ischemia, cerebral monoamines and hydroxyl radical production overloads, and improves neuronal damage and the ST in heat stroke rats. Our data suggest that administration of this combined agent seems to have more effective to ameliorate the heat stroke-induced neuronal damage and prolong the ST.


Assuntos
Dexametasona/administração & dosagem , Golpe de Calor/tratamento farmacológico , Manitol/administração & dosagem , Neurônios/efeitos dos fármacos , Neurônios/patologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Quimioterapia Combinada , Golpe de Calor/metabolismo , Golpe de Calor/patologia , Masculino , Neurônios/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA