Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Am J Respir Cell Mol Biol ; 71(2): 195-206, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38597725

RESUMEN

Extreme heat caused by climate change is increasing the transmission of infectious diseases, resulting in a sharp rise in heat-related illness and mortality. Understanding the mechanistic link between heat, inflammation, and disease is thus important for public health. Thermal hyperpnea, and consequent respiratory alkalosis, is crucial in febrile seizures and convulsions induced by heat stress in humans. Here, we address what causes thermal hyperpnea in neonates and how it is affected by inflammation. Transient receptor potential cation channel subfamily V member 1 (TRPV1), a heat-activated channel, is sensitized by inflammation and modulates breathing and thus may play a key role. To investigate whether inflammatory sensitization of TRPV1 modifies neonatal ventilatory responses to heat stress, leading to respiratory alkalosis and an increased susceptibility to hyperthermic seizures, we treated neonatal rats with bacterial LPS, and breathing, arterial pH, in vitro vagus nerve activity, and seizure susceptibility were assessed during heat stress in the presence or absence of a TRPV1 antagonist (AMG-9810) or shRNA-mediated TRPV1 suppression. LPS-induced inflammatory preconditioning lowered the threshold temperature and latency of hyperthermic seizures. This was accompanied by increased tidal volume, minute ventilation, expired CO2, and arterial pH (alkalosis). LPS exposure also elevated vagal spiking and intracellular calcium concentrations in response to hyperthermia. TRPV1 inhibition with AMG-9810 or shRNA reduced the LPS-induced susceptibility to hyperthermic seizures and altered the breathing pattern to fast shallow breaths (tachypnea), making each breath less efficient and restoring arterial pH. These results indicate that inflammation exacerbates thermal hyperpnea-induced respiratory alkalosis associated with increased susceptibility to hyperthermic seizures, primarily mediated by TRPV1 localized to vagus neurons.


Asunto(s)
Inflamación , Convulsiones Febriles , Canales Catiónicos TRPV , Convulsiones Febriles/fisiopatología , Convulsiones Febriles/metabolismo , Animales , Canales Catiónicos TRPV/metabolismo , Inflamación/metabolismo , Ratas , Respuesta al Choque Térmico , Animales Recién Nacidos , Lipopolisacáridos/farmacología , Nervio Vago/fisiopatología , Ratas Sprague-Dawley , Alcalosis Respiratoria/metabolismo , Alcalosis Respiratoria/fisiopatología , Hipertermia/metabolismo , Hipertermia/fisiopatología
2.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33972431

RESUMEN

Febrile seizures (FSs) are the most common convulsion in infancy and childhood. Considering the limitations of current treatments, it is important to examine the mechanistic cause of FSs. Prompted by a genome-wide association study identifying TMEM16C (also known as ANO3) as a risk factor of FSs, we showed previously that loss of TMEM16C function causes hippocampal neuronal hyperexcitability [Feenstra et al., Nat. Genet. 46, 1274-1282 (2014)]. Our previous study further revealed a reduction in the number of warm-sensitive neurons that increase their action potential firing rate with rising temperature of the brain region harboring these hypothalamic neurons. Whereas central neuronal hyperexcitability has been implicated in FSs, it is unclear whether the maximal temperature reached during fever or the rate of body temperature rise affects FSs. Here we report that mutant rodent pups with TMEM16C eliminated from all or a subset of their central neurons serve as FS models with deficient thermoregulation. Tmem16c knockout (KO) rat pups at postnatal day 10 (P10) are more susceptible to hyperthermia-induced seizures. Moreover, they display a more rapid rise of body temperature upon heat exposure. In addition, conditional knockout (cKO) mouse pups (P11) with TMEM16C deletion from the brain display greater susceptibility of hyperthermia-induced seizures as well as deficiency in thermoregulation. We also found similar phenotypes in P11 cKO mouse pups with TMEM16C deletion from Ptgds-expressing cells, including temperature-sensitive neurons in the preoptic area (POA) of the anterior hypothalamus, the brain region that controls body temperature. These findings suggest that homeostatic thermoregulation plays an important role in FSs.


Asunto(s)
Regulación de la Temperatura Corporal/genética , Canales de Cloruro/genética , Fiebre/genética , Hipertermia/genética , Área Preóptica/metabolismo , Convulsiones Febriles/genética , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/fisiología , Canales de Cloruro/deficiencia , Femenino , Fiebre/inducido químicamente , Fiebre/metabolismo , Fiebre/fisiopatología , Expresión Génica , Hipocampo/metabolismo , Hipocampo/fisiopatología , Hipertermia/metabolismo , Hipertermia/fisiopatología , Ácido Kaínico/administración & dosificación , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Área Preóptica/fisiopatología , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Ratas , Convulsiones Febriles/inducido químicamente , Convulsiones Febriles/metabolismo , Convulsiones Febriles/fisiopatología
3.
Int J Mol Sci ; 25(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39201559

RESUMEN

In order to improve naturally occurring xanthones' anticancer properties, chemical synthesis is proposed. In this study, from eight novel xanthone derivatives coupled to morpholine or aminoalkyl morpholine, only the two most active ones were chosen. For additional enhancement of the anticancer activity of our tested compounds, we combined chemotherapy with hyperthermia in the range of 39-41 °C, from which the mild conditions of 39 °C were the most influencing. This approach had a profound impact on the anticancer properties of the tested compounds. TOV-21G and SC-OV-3 ovarian cell line motility and metastasis behavior were tested in native and hyperthermia conditions, indicating decreased wound healing properties and clonogenic activity. Similarly, the expression of genes involved in metastasis was hampered. The expression of heat shock proteins involved in cancer progression (Hsc70, HSP90A, and HSP90B) was significantly influenced by xanthone derivatives. Chemotherapy in mild hyperthermia conditions had also an impact on decreasing mitochondria potential, visualized with JC-1. Synthetic xanthone ring modifications may increase the anticancer activity of the obtained substances. Additional improvement of their activity can be achieved by applying mild hyperthermia conditions. Further development of a combined anticancer therapy approach may result in increasing currently known chemotherapeutics, resulting in a greater recovery rate and diminishment of the cytotoxicity of drugs.


Asunto(s)
Antineoplásicos , Xantonas , Xantonas/farmacología , Xantonas/química , Humanos , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Movimiento Celular/efectos de los fármacos , Hipertermia Inducida/métodos , Femenino , Hipertermia/tratamiento farmacológico , Hipertermia/metabolismo
4.
Am J Physiol Regul Integr Comp Physiol ; 325(6): R725-R734, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37811714

RESUMEN

Exercise is associated with the development of oxidative stress, but the specific source and mechanism of production of pro-oxidant chemicals during exercise has not been confirmed. We used equine skeletal muscle mitochondria to test the hypothesis that hyperthermia and acidosis affect mitochondrial oxygen consumption and production of reactive oxygen species (ROS). Skeletal muscle biopsies were obtained at rest, after an acute episode of fatiguing exercise, and after a 9-wk conditioning program to increase aerobic fitness. Mitochondrial oxygen consumption and ROS production were measured simultaneously using high-resolution respirometry. Both hyperthermia and acidosis increased nonphosphorylating (LEAK) respiration (5.8× and 3.0×, respectively, P < 0.001) and decreased efficiency of oxidative phosphorylation. The combined effects of hyperthermia and acidosis resulted in large decreases in phosphorylating respiration, further decreasing oxidative phosphorylation efficiency from 97% to 86% (P < 0.01). Increased aerobic fitness reduced the effects of acidosis on LEAK respiration. Hyperthermia increased and acidosis decreased ROS production (2× and 0.23×, respectively, P < 0.001). There was no effect of acute exercise, but an aerobic conditioning program was associated with increased ROS production during both nonphosphorylating and phosphorylating respiration. Hyperthermia increased the ratio of ROS production to O2 consumption during phosphorylating respiration, suggesting that high-temperature impaired transfer of energy through the electron transfer system despite relatively low mitochondrial membrane potential. These data support the role of skeletal muscle mitochondria in the development of exercise-induced oxidative stress, particularly during forms of exercise that result in prolonged hyperthermia without acidosis.NEW & NOTEWORTHY The results of this study provide evidence for the role of mitochondria-derived ROS in the development of systemic oxidative stress during exercise as well as skeletal muscle diseases such as exertional rhabdomyolysis.


Asunto(s)
Acidosis , Hipertermia Inducida , Animales , Caballos , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Acidosis/metabolismo , Consumo de Oxígeno/fisiología , Hipertermia/metabolismo
5.
Am J Physiol Endocrinol Metab ; 322(1): E10-E23, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34779255

RESUMEN

Cholecystokinin (CCK) increases core body temperature via CCK2 receptors when administered intracerebroventricularly (icv). The mechanisms of CCK-induced hyperthermia are unknown, and it is also unknown whether CCK contributes to the fever response to systemic inflammation. We studied the interaction between central CCK signaling and the cyclooxygenase (COX) pathway. Body temperature was measured in adult male Wistar rats pretreated with intraperitoneal infusion of the nonselective COX enzyme inhibitor metamizol (120 mg/kg) or a selective COX-2 inhibitor, meloxicam, or etoricoxib (10 mg/kg for both) and, 30 min later, treated with intracerebroventricular CCK (1.7 µg/kg). In separate experiments, CCK-induced neuronal activation (with and without COX inhibition) was studied in thermoregulation- and feeding-related nuclei with c-Fos immunohistochemistry. CCK increased body temperature by ∼0.4°C from 10 min postinfusion, which was attenuated by metamizol. CCK reduced the number of c-Fos-positive cells in the median preoptic area (by ∼70%) but increased it in the dorsal hypothalamic area and in the rostral raphe pallidus (by ∼50% in both); all these changes were completely blocked with metamizol. In contrast, CCK-induced satiety and neuronal activation in the ventromedial hypothalamus were not influenced by metamizol. CCK-induced hyperthermia was also completely blocked with both selective COX-2 inhibitors studied. Finally, the CCK2 receptor antagonist YM022 (10 µg/kg icv) attenuated the late phases of fever induced by bacterial lipopolysaccharide (10 µg/kg; intravenously). We conclude that centrally administered CCK causes hyperthermia through changes in the activity of "classical" thermoeffector pathways and that the activation of COX-2 is required for the development of this response.NEW & NOTEWORTHY An association between central cholecystokinin signaling and the cyclooxygenase-prostaglandin E pathway has been proposed but remained poorly understood. We show that the hyperthermic response to the central administration of cholecystokinin alters the neuronal activity within efferent thermoeffector pathways and that these effects are fully blocked by the inhibition of cyclooxygenase. We also show that the activation of cyclooxygenase-2 is required for the hyperthermic effect of cholecystokinin and that cholecystokinin is a modulator of endotoxin-induced fever.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Colecistoquinina/administración & dosificación , Ciclooxigenasa 2/metabolismo , Hipertermia/inducido químicamente , Hipertermia/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Anorexia/inducido químicamente , Benzodiazepinas/administración & dosificación , Regulación de la Temperatura Corporal/efectos de los fármacos , Colecistoquinina/efectos adversos , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Fiebre/inducido químicamente , Fiebre/tratamiento farmacológico , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Lipopolisacáridos/efectos adversos , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Receptor de Colecistoquinina B/antagonistas & inhibidores , Resultado del Tratamiento
6.
Biochem J ; 478(1): 179-196, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33346336

RESUMEN

Human body temperature limits below 40°C during heat stroke or fever. The implications of prolonged exposure to the physiologically relevant temperature (40°C) on cellular mechanobiology is poorly understood. Here, we have examined the effects of heat stress (40°C for 72 h incubation) in human lung adenocarcinoma (A549), mouse melanoma (B16F10), and non-cancerous mouse origin adipose tissue cells (L929). Hyperthermia increased the level of ROS, γ-H2AX and HSP70 and decreased mitochondrial membrane potential in the cells. Heat stress impaired cell division, caused G1 arrest, induced cellular senescence, and apoptosis in all the tested cell lines. The cells incubated at 40°C for 72 h displayed a significant decrease in the f-actin level and cellular traction as compared with cells incubated at 37°C. Also, the cells showed a larger focal adhesion area and stronger adhesion at 40°C than at 37°C. The mitotic cells at 40°C were unable to round up properly and displayed retracting actin stress fibers. Hyperthermia down-regulated HDAC6, increased the acetylation level of microtubules, and perturbed the chromosome alignment in the mitotic cells at 40°C. Overexpression of HDAC6 rescued the cells from the G1 arrest and reduced the delay in cell rounding at 40°C suggesting a crucial role of HDAC6 in hyperthermia mediated responses. This study elucidates the significant role of cellular traction, focal adhesions, and cytoskeletal networks in mitotic cell rounding and chromosomal misalignment. It also highlights the significance of HDAC6 in heat-evoked senile cellular responses.


Asunto(s)
Senescencia Celular/fisiología , Citoesqueleto/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Histona Desacetilasa 6/metabolismo , Hipertermia/metabolismo , Fibras de Estrés/metabolismo , Acetilación , Actinas/metabolismo , Apoptosis/fisiología , Adhesión Celular/fisiología , División Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Forma de la Célula , Tamaño de la Célula , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Puntos de Control de la Fase G1 del Ciclo Celular/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Histona Desacetilasa 6/genética , Humanos , Hipertermia/genética , Potencial de la Membrana Mitocondrial/fisiología , Microtúbulos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
7.
J Biol Chem ; 295(45): 15226-15235, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-32826313

RESUMEN

Mutations in the skeletal muscle ryanodine receptor gene (RYR1) can cause susceptibility to malignant hyperthermia (MH), a potentially lethal genetic condition triggered by volatile anesthetics. MH is associated with hypermetabolism, which has directed research interest into oxidative phosphorylation and muscle bioenergetics. The most common cause of MH in the United Kingdom is the c.7300G>A RYR1 variant, which is present in ∼16% of MH families. Our study focuses on the MH susceptible G2435R-RYR1 knock-in mouse model, which is the murine equivalent of the human c.7300G>A genotype. Using a combination of transcriptomics, protein expression, and functional analysis, we investigated adult muscle fiber bioenergetics in this mouse model. RNA-Seq data showed reduced expression of genes associated with mitochondria and fatty acid oxidation in RYR1 mutants when compared with WT controls. Mitochondrial function was assessed by measuring oxygen consumption rates in permeabilized muscle fibers. Comparisons between WT and homozygous G2435R-RYR1 mitochondria showed a significant increase in complex I-facilitated oxidative phosphorylation in mutant muscle. Furthermore, we observed a gene-dose-specific increase in reactive oxygen species production in G2435R-RYR1 muscle fibers. Collectively, these findings provide evidence of metabolic defects in G2435R-RYR1 knock-in mouse muscle under basal conditions. Differences in metabolic profile could be the result of differential gene expression in metabolic pathways, in conjunction with mitochondrial damage accumulated from chronic exposure to increased oxidative stress.


Asunto(s)
Hipertermia/genética , Hipertermia/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Femenino , Masculino , Ratones
8.
J Neurochem ; 156(5): 604-613, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32858780

RESUMEN

De novo heterozygous mutations in the STX1B gene, encoding syntaxin 1B, cause a familial, fever-associated epilepsy syndrome. Syntaxin 1B is an essential component of the pre-synaptic neurotransmitter release machinery as a soluble N-ethylmaleimide-sensitive factor attachment protein receptor protein that regulates the exocytosis of synaptic vesicles. It is also involved in regulating the functions of the SLC6 family of neurotransmitter transporters that reuptake neurotransmitters, including inhibitory neurotransmitters, such as γ-aminobutyric acid (GABA) and glycine. The purpose of the present study was to elucidate the molecular mechanisms underlying the development of febrile seizures by examining the effects of syntaxin 1B haploinsufficiency on inhibitory synaptic transmission during hyperthermia in a mouse model. Stx1b gene heterozygous knockout (Stx1b+/- ) mice showed increased susceptibility to febrile seizures and drug-induced seizures. In cultured hippocampal neurons, we examined the temperature-dependent properties of neurotransmitter release and reuptake by GABA transporter-1 (GAT-1) at GABAergic neurons using whole-cell patch-clamp recordings. The rate of spontaneous quantal GABA release was reduced in Stx1b+/- mice. The hyperthermic temperature increased the tonic GABAA current in wild-type (WT) synapses, but not in Stx1b+/- synapses. In WT neurons, recurrent bursting activities were reduced in a GABA-dependent manner at hyperthermic temperature; however, this was abolished in Stx1b+/- neurons. The blockade of GAT-1 increased the tonic GABAA current and suppressed recurrent bursting activities in Stx1b+/- neurons at the hyperthermic temperature. These data suggest that functional abnormalities associated with GABA release and reuptake in the pre-synaptic terminals of GABAergic neurons may increase the excitability of the neural circuit with hyperthermia.


Asunto(s)
Temperatura Corporal/fisiología , Líquido Extracelular/metabolismo , Convulsiones/metabolismo , Sinapsis/metabolismo , Sintaxina 1/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Hipocampo/metabolismo , Hipertermia/genética , Hipertermia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pentilenotetrazol/toxicidad , Convulsiones/inducido químicamente , Convulsiones/genética , Sinapsis/genética , Sintaxina 1/genética
9.
Am J Physiol Regul Integr Comp Physiol ; 320(4): R563-R573, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33085914

RESUMEN

Transient receptor potential vanilloid 4 (TRPV4) channels exist on vascular endothelial cells and eccrine sweat gland secretory cells in human skin. Here, we assessed whether TRPV4 channels contribute to cutaneous vasodilation and sweating during whole body passive heat stress (protocol 1) and to cutaneous vasodilation during postocclusive reactive hyperemia and local thermal hyperemia (protocol 2). Intradermal microdialysis was employed to locally deliver pharmacological agents to forearm skin sites, where cutaneous vascular conductance (CVC) and sweat rate were assessed. In protocol 1 (12 young adults), CVC and sweat rate were increased by passive whole body heating, resulting in a body core temperature elevation of 1.2 ± 0.1°C. The elevated CVC and sweat rate assessed at sites treated with TRPV4 channel antagonist (either 200 µM HC-067047 or 125 µM GSK2193874) were not different from the vehicle control site (5% dimethyl sulfoxide). After whole body heating, the TRPV4 channel agonist (100 µM GSK1016790A) was administered to each skin site, eliciting elevations in CVC. Relative to control, this response was partly attenuated by both TRPV4 channel antagonists, confirming drug efficacy. In protocol 2 (10 young adults), CVC was increased following a 5-min arterial occlusion and during local heating from 33 to 42°C. These responses did not differ between the control and the TRPV4 channel antagonist sites (200 µM HC-067047). We show that TRPV4 channels are not required for regulating cutaneous vasodilation or sweating during a whole body passive heat stress. Furthermore, they are not required for regulating cutaneous vasodilation during postocclusive reactive hyperemia and local thermal hyperemia.


Asunto(s)
Hiperemia/fisiopatología , Hipertermia/fisiopatología , Moduladores del Transporte de Membrana/administración & dosificación , Piel/irrigación sanguínea , Sudoración , Canales Catiónicos TRPV/antagonistas & inhibidores , Vasodilatación , Adulto , Femenino , Humanos , Hiperemia/metabolismo , Hipertermia/metabolismo , Leucina/administración & dosificación , Leucina/análogos & derivados , Masculino , Microdiálisis , Morfolinas/administración & dosificación , Piperidinas/administración & dosificación , Pirroles/administración & dosificación , Quinolinas/administración & dosificación , Flujo Sanguíneo Regional , Piel/metabolismo , Sulfonamidas/administración & dosificación , Canales Catiónicos TRPV/metabolismo , Factores de Tiempo , Adulto Joven
10.
FASEB J ; 34(6): 8721-8733, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32367593

RESUMEN

Malignant hyperthermia (MH) is characterized by induction of skeletal muscle hyperthermia in response to a dysregulated increase in myoplasmic calcium. Although altered energetics play a central role in MH, MH-susceptible humans and mouse models are often described as having no phenotype until exposure to a triggering agent. The purpose of this study was to determine the influence of the R163C ryanodine receptor 1 mutation, a common MH mutation in humans, on energy expenditure, and voluntary wheel running in mice. Energy expenditure was measured by indirect respiration calorimetry in wild-type (WT) and heterozygous R163C (HET) mice over a range of ambient temperatures. Energy expenditure adjusted for body weight or lean mass was increased (P < .05) in male, but not female, HET mice housed at 22°C or when housed at 28°C with a running wheel. In female mice, voluntary wheel running was decreased (P < .05) in the HET vs WT animals when analyzed across ambient temperatures. The thermoneutral zone was also widened in both male and female HET mice. The results of the study show that the R163C mutations alters energetics even at temperatures that do not typically induce MH.


Asunto(s)
Metabolismo Energético/fisiología , Hipertermia/patología , Hipertermia Maligna/patología , Actividad Motora/fisiología , Animales , Calcio/metabolismo , Señalización del Calcio/fisiología , Femenino , Heterocigoto , Hipertermia/metabolismo , Masculino , Hipertermia Maligna/metabolismo , Potenciales de la Membrana/fisiología , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación/genética , Canal Liberador de Calcio Receptor de Rianodina/genética
11.
J Nanobiotechnology ; 19(1): 210, 2021 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-34261493

RESUMEN

BACKGROUND: We investigated the therapeutic effect of targeting extracellular vesicles (EVs) loaded with indocyanine green (ICG) and paclitaxel (PTX) on glioma. METHODS: Raw264.7 cells were harvested to extract EVs for the preparation of ICG/PTX@RGE-EV by electroporation and click chemistry. We evaluated the success of modifying Neuropilin-1 targeting peptide (RGE) on the EV membrane of ICG/PTX@RGE-EV using super-resolution fluorescence microscopy and flow cytometry. Spectrophotometry and high performance liquid chromatography (HPLC) were implemented for qualitative and quantitative analysis of the ICG and PTX loaded in EVs. Photothermal properties of the vesicles were evaluated by exposing to 808-nm laser light. Western blot analysis, cell counting kit 8 (CCK-8), Calcein Acetoxymethyl Ester/propidium iodide (Calcein-AM/PI) staining, and flow cytometry were utilized for assessing effects of vesicle treatment on cellular behaviors. A nude mouse model bearing glioma was established to test the targeting ability and anti-tumor action of ICG/PTX@RGE-EV in vivo. RESULTS: Under exposure to 808-nm laser light, ICG/PTX@RGE-EV showed good photothermal properties and promotion of PTX release from EVs. ICG/PTX@RGE-EV effectively targeted U251 cells, with activation of the Caspase-3 pathway and elevated apoptosis in U251 cells through chemotherapy combined with hyperthermia. The anti-tumor function of ICG/PTX@RGE-EV was confirmed in the glioma mice via increased accumulation of PTX in the ICG/PTX@RGE-EV group and an increased median survival of 48 days in the ICG/PTX@RGE-EV group as compared to 25 days in the PBS group. CONCLUSION: ICG/PTX@RGE-EV might actively target glioma to repress tumor growth by accelerating glioma cell apoptosis through combined chemotherapy-hyperthermia.


Asunto(s)
Biomimética/métodos , Vesículas Extracelulares/efectos de los fármacos , Glioma/tratamiento farmacológico , Hipertermia/tratamiento farmacológico , Verde de Indocianina/química , Rayos Infrarrojos , Nanopartículas/química , Imagen Óptica/métodos , Paclitaxel/farmacología , Animales , Caspasa 3 , Línea Celular Tumoral , Quimioterapia/métodos , Fluorescencia , Glioma/patología , Humanos , Hipertermia/diagnóstico por imagen , Hipertermia/metabolismo , Hipertermia/patología , Ratones , Ratones Desnudos , Neuropilina-1 , Células RAW 264.7
12.
Int J Sports Med ; 42(8): 673-681, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33772503

RESUMEN

The active participation of skeletal muscles is a unique characteristic of exertional heat stroke. Nevertheless, the only well-documented link between skeletal muscle activities and exertional heat stroke pathophysiology is the extensive muscle damage (e. g., rhabdomyolysis) and subsequent leakage of intramuscular content into the circulation of exertional heat stroke victims. Here, we will present and discuss rarely explored roles of skeletal muscles in the context of exertional heat stroke pathophysiology and recovery. This includes an overview of heat production that contributes to severe hyperthermia and the synthesis and secretion of bioactive molecules, such as cytokines, chemokines and acute phase proteins. These molecules can alter the overall inflammatory status from pro- to anti-inflammatory, affecting other organ systems and influencing recovery. The activation of innate immunity can determine whether a victim is ready to return to physical activity or experiences a prolonged convalescence. We also provide a brief discussion on whether heat acclimation can shift skeletal muscle secretory phenotype to prevent or aid recovery from exertional heat stroke. We conclude that skeletal muscles should be considered as a key organ system in exertional heat stroke pathophysiology.


Asunto(s)
Golpe de Calor/fisiopatología , Músculo Esquelético/fisiopatología , Esfuerzo Físico/fisiología , Aclimatación/fisiología , Proteínas de Fase Aguda/metabolismo , Calcio/metabolismo , Quimiocinas/metabolismo , Convalecencia , Citocinas/metabolismo , Agotamiento por Calor , Golpe de Calor/sangre , Golpe de Calor/etiología , Golpe de Calor/inmunología , Humanos , Hipertermia/etiología , Hipertermia/metabolismo , Hipertermia/fisiopatología , Inmunidad Innata/fisiología , Contracción Muscular/fisiología , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Esfuerzo Físico/inmunología , Recuperación de la Función , Rabdomiólisis/etiología , Termogénesis/fisiología , Termotolerancia/fisiología
13.
J Therm Biol ; 99: 102943, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34420607

RESUMEN

BACKGROUND: Hyperthermia, induced by exercise in the heat, alters the redox status. The physiological significance of these observations remains uncertain but may justify why the consequences of exercising in the heat span from positive health adaptations to negative and even lethal outcomes. Here, we conducted a systematic review to investigate the redox responses during acute exercise in the heat in healthy adults. METHODS: We searched MEDLINE, Cochrane Wiley, ClinicalTrials.gov, PEDRO and LILACS for clinical trials investigating pro- and antioxidant responses to exercise associated with hyperthermia and/or sweat-induced dehydration in healthy young individuals. Two independent reviewers extracted data and assessed the quality of the included studies. RESULTS: A total of 1,014 records were selected, nine full papers were evaluated for eligibility, and eight studies met the inclusion criteria. Overall, results show that hyperthermia promotes oxidative stress both at the tissue level and in the circulation. Exercising in the heat heightens endogenous antioxidant defense systems, attenuating the negative effects of hyperthermia on oxidative damage. Studies also indicate that sweat-induced dehydration promotes oxidative stress, which is attenuated by rehydration. CONCLUSION: These findings suggest that changes in redox status play a role in determining whether an acute bout of exercise in the heat lead to adaptive or maladaptive outcomes.


Asunto(s)
Ejercicio Físico/fisiología , Hipertermia/metabolismo , Estrés Oxidativo , Antioxidantes/metabolismo , Deshidratación/metabolismo , Humanos , Oxidación-Reducción
14.
Nanomedicine ; 28: 102183, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32222478

RESUMEN

Desmoplasia, an aberrant production of extracellular matrix (ECM), is considered as one predictive marker of malignancy of pancreatic cancer. In this paper, we study the effect of mild hyperthermia on fibrillary collagen architecture in murine Achilles tendons and in a pancreatic cancer model, in vitro, i.e. 3D hetero-type tumor spheroids, consisting of pancreatic cancer (Panc-1) cells and fibroblasts (WI-38), producing collagen fibers. We clearly demonstrate that i) mild hyperthermia (40 °C, 42 °C) damages the collagen architecture in murine Achilles tendons. ii) Mild extrinsic (hot air) and iron oxide nanoparticle based magnetic hyperthermia reduce the level of collagen fiber architecture in the generated hetero-type tumor spheroids. iii) Mild magnetic hyperthermia reduces cell vitality mainly through apoptotic and necrotic processes in the generated tumor spheroids. In conclusion, hetero-type 3D tumor spheroids are suitable for studying the effect of hyperthermia on collagen fibers, in vitro.


Asunto(s)
Colágeno/metabolismo , Hipertermia/metabolismo , Neoplasias Pancreáticas/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Humanos , Nanopartículas Magnéticas de Óxido de Hierro/química , Ratones , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
15.
J Therm Biol ; 91: 102610, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32716860

RESUMEN

We investigated whether the magnitude of exercise-induced hyperthermia influences intestinal permeability and tight junction gene expression. Twenty-nine male Wistar rats were divided into four groups: rest at 24 °C and exercise at 13 °C, 24 °C or 31 °C. The exercise consisted of a 90-min treadmill run at 15 m/min, and different ambient temperatures were used to produce distinct levels of exercise-induced hyperthermia. Before the experimental trials, the rats were treated by gavage with diethylenetriaminepentaacetic acid labeled with technetium-99 metastable as a radioactive probe. The rats' core body temperature (TCORE) was measured by telemetry. Immediately after the trials, the rats were euthanized, and the intestinal permeability was assessed by measuring the radioactivity of blood samples. The mRNA levels of occludin and zonula occludens-1 (ZO-1) genes were determined in duodenum samples. Exercise at 24 °C increased TCORE to values close to 39 °C, without changing permeability compared with the resting trial at the same environment. Meanwhile, rats' TCORE exceeded 40 °C during exercise at 31 °C, leading to greater permeability relative to those observed after exercise in the other ambient temperatures (e.g., 0.0037%/g at 31 °C vs. 0.0005%/g at 13 °C; data expressed as medians; p < 0.05). Likewise, the rats exercised at 31 °C exhibited higher mRNA levels of ZO-1 and occludin genes than the rats exercised at 24 °C or 13 °C. The changes in permeability and gene expression were positively and significantly associated with the magnitude of hyperthermia. We conclude that marked hyperthermia caused by exercise in the warmer environment increases intestinal permeability and mRNA levels of tight junction genes.


Asunto(s)
Hipertermia/metabolismo , Mucosa Intestinal/metabolismo , Ocludina/genética , Esfuerzo Físico , Proteína de la Zonula Occludens-1/genética , Animales , Hipertermia/etiología , Absorción Intestinal , Masculino , Ocludina/metabolismo , Ratas , Ratas Wistar , Proteína de la Zonula Occludens-1/metabolismo
16.
J Therm Biol ; 91: 102615, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32716865

RESUMEN

Protein homeostasis or proteostasis, the correct balance between production and degradation of proteins, is an essential pillar for proper cellular function. Among the several cellular mechanisms that disrupt homeostatic conditions in cancer cells, hyperthermia (HT) has shown promising anti-tumor effects. However, cancer cells are also capable of thermoresistance. Indeed, HT-induced protein denaturation and aggregation results in the up regulation of heat shock proteins, a group of molecular chaperones with cytoprotective and anti-apoptotic properties via stress-inducible transcription factor, heat shock factor 1(HSF1). Heat shock proteins assist in the refolding of misfolded proteins and aids in their elimination if they become irreversibly damaged by various stressors. Furthermore, HSF1 also initiates the unfolded protein response in the endoplasmic reticulum (ER) to assist in the protein folding capacity of ER and also promotes the translation of pro-survival proteins' mRNA such as activating transcription factor 4 (ATF 4). Moreover, HT associated induction of microRNAs is also involved in thermal resistance of cancer cells via up-regulation of anti-apoptotic Bcl-2 proteins and down regulation of pro-apoptotic Bax and caspase 3 activities. Another cellular protection in response to stressors is Autophagy, which is regulated by the Mammalian target of rapamycin (mTOR) protein. Kinase activity in mTOR phosphorylates HSF1 and promotes its nuclear translocation for heat shock protein synthesis. Over-expression of heat shock proteins are reported to up-regulate Beclin-1, an autophagy initiator. Moreover, HT-induced reactive oxygen species (ROS) generation is sensitized by transcription factor NF-E2 related factor 2 (Nrf2) and activates the cellular expression of antioxidants and autophagy gene. Furthermore, ROS also potentiates autophagy via activation of Beclin-1. Inhibition of thermotolerance can potentiate HT-induced apoptosis. Here, we outlined that heat stress alters cellular proteins which activates cellular homeostatic processes to promote cell survival and make cancer cells thermotolerant.


Asunto(s)
Apoptosis , Hipertermia Inducida/efectos adversos , Hipertermia/metabolismo , Proteostasis , Animales , Autofagia , Respuesta al Choque Térmico , Humanos , Hipertermia/etiología , Estrés Oxidativo
17.
J Therm Biol ; 91: 102619, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32716869

RESUMEN

The objective of this study was to examine the effects of chronic cyclic heat stress (HS) on the intestinal morphology, oxidative stress and cecal bacterial communities of broilers. One-day-old Arbor Acres (AA) male broilers (n = 100) were acclimated for 3 weeks and then randomly allocated into two groups, normal control (NC) group (22 ± 1 °C, 24 h/day) and HS group (32 ± 1 °C, 10 h/day lasted for 2 weeks). At 35 d of age, intestinal segments (duodenum, jejunum and ileum) and cecal digesta were collected for detection. HS affected intestinal morphology, inducing epithelial cell abscission, inflammatory cell infiltration, and lamina propria edema. Compared with the NC group, HS significantly decreased (P < 0.01) villus height (VH) and the VH-to-crypt depth (CD) ratio (VCR), increased (P < 0.05) CD in the duodenum and ileum, but had no effect on the VH in the jejunum. Moreover, HS induced oxidative stress with antioxidant enzymes activity decreasing (P < 0.05) while malondialdehyde (MDA) content increasing (P < 0.05) in small intestine. Pearson's correlation analysis indicated that MDA content was negatively correlated with VH (P < 0.05). The result of 16S rRNA sequencing showed that HS exposure impacted cecal microbiota alpha diversity (phylogenetic diversity whole-tree index) and beta diversity. Based on principal coordinate analysis (PCoA) plots for weighted UniFrac metrics and unweighted pair group method with arithmetic mean (UPGMA), there were 8 discriminative features at the genus level (linear discriminant analysis score > 2). Parabacteroides, Saccharimonas, Romboutsia and Weissella were reduced, while Anaerofustis, Pseudonocardia, Rikenella and Tyzzerella were enriched in heat-stressed broilers. Collectively, these results indicated that chronic cyclic HS induced oxidative stress that caused damage to intestinal villus-crypt structures, and then altered the cecal microflora profile.


Asunto(s)
Pollos/fisiología , Microbioma Gastrointestinal , Respuesta al Choque Térmico , Hipertermia/veterinaria , Intestino Delgado/metabolismo , Animales , Ciego/microbiología , Pollos/metabolismo , Pollos/microbiología , Hipertermia/metabolismo , Hipertermia/microbiología , Intestino Delgado/microbiología , Intestino Delgado/patología , Masculino , Estrés Oxidativo
18.
ACS Biomater Sci Eng ; 10(5): 2995-3005, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38654432

RESUMEN

Magnetic hyperthermia is a crucial medical engineering technique for treating diseases, which usually uses alternating magnetic fields (AMF) to interplay with magnetic substances to generate heat. Recently, it has been found that in some cases, there is no detectable temperature increment after applying an AMF, which caused corresponding effects surprisingly. The mechanisms involved in this phenomenon are not yet fully understood. In this study, we aimed to explore the role of Ca2+ overload in the magnetic hyperthermia effect without a perceptible temperature rise. A cellular system expressing the fusion proteins TRPV1 and ferritin was prepared. The application of an AMF (518 kHz, 16 kA/m) could induce the fusion protein to release a large amount of iron ions, which then participates in the production of massive reactive oxygen radicals (ROS). Both ROS and its induced lipid oxidation enticed the opening of ion channels, causing intracellular Ca2+ overload, which further led to decreased cellular viability. Taken together, Ca2+ overload triggered by elevated ROS and the induced oxidation of lipids contributes to the magnetic hyperthermia effect without a perceptible temperature rise. These findings would be beneficial for expanding the application of temperature-free magnetic hyperthermia, such as in cellular and neural regulation, design of new cancer treatment methods.


Asunto(s)
Calcio , Supervivencia Celular , Hipertermia Inducida , Campos Magnéticos , Especies Reactivas de Oxígeno , Canales Catiónicos TRPV , Calcio/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPV/metabolismo , Humanos , Hipertermia Inducida/métodos , Temperatura , Ferritinas/metabolismo , Hipertermia/metabolismo
19.
Cells ; 13(13)2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38994992

RESUMEN

Previous studies reported that a mild, non-protein-denaturing, fever-like temperature increase induced the unfolded protein response (UPR) in mammalian cells. Our dSTORM super-resolution microscopy experiments revealed that the master regulator of the UPR, the IRE1 (inositol-requiring enzyme 1) protein, is clustered as a result of UPR activation in a human osteosarcoma cell line (U2OS) upon mild heat stress. Using ER thermo yellow, a temperature-sensitive fluorescent probe targeted to the endoplasmic reticulum (ER), we detected significant intracellular thermogenesis in mouse embryonic fibroblast (MEF) cells. Temperatures reached at least 8 °C higher than the external environment (40 °C), resulting in exceptionally high ER temperatures similar to those previously described for mitochondria. Mild heat-induced thermogenesis in the ER of MEF cells was likely due to the uncoupling of the Ca2+/ATPase (SERCA) pump. The high ER temperatures initiated a pronounced cytosolic heat-shock response in MEF cells, which was significantly lower in U2OS cells in which both the ER thermogenesis and SERCA pump uncoupling were absent. Our results suggest that depending on intrinsic cellular properties, mild hyperthermia-induced intracellular thermogenesis defines the cellular response mechanism and determines the outcome of hyperthermic stress.


Asunto(s)
Retículo Endoplásmico , Respuesta al Choque Térmico , Termogénesis , Humanos , Animales , Retículo Endoplásmico/metabolismo , Ratones , Respuesta de Proteína Desplegada , Línea Celular Tumoral , Estrés del Retículo Endoplásmico , Hipertermia/metabolismo , Hipertermia/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
20.
J Tradit Chin Med ; 44(5): 954-962, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39380226

RESUMEN

OBJECTIVE: To explore how Qingfei Zhisou oral liquid (, QFZS) adjusts body temperature bias and the interaction of inflammatory factors levels and metabolomic differences. METHODS: Dry yeast was subcutaneously injected at 10 mL/kg to establish the pyrexia model. We randomly divided 60 Sprague-Dawley rats into five groups: control, model, positive, low dose of QFZS and high dose of QFZS. Inflammatory proteins were evaluated by Western blotting and immunohistochemistry. For the examination of the endogenous metabolites, enzyme linked immunosorbent assay and ultra-high-performance liquid chromatography high-resolution mass spectrometry were employed. RESULTS: QFZS significantly reduced rats' body temperature within 6 h after dry yeast injection and reduced the secretion of the arginine vasopressin, cyclic adenosine monophosphate, prostaglandin E-2, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1ß in serum. Meanwhile, we identified 41 metabolites between the model and QFZS groups, including arachidonic acid and lysophospholipids. QFZS restored normal arachidonic acid levels. Based on the differential metabolite enrichment analysis, QFZS's anti-inflammatory and anti-pyrexia effects might be related to the inflammatory pathway regulated by transient receptor potential. Additionally, QFZS treatment reduced transient receptor potential melastatin 2 ion channel expression and affected TNF-α, heat shock protein 70, and cyclooxygenase-2 expression in the hypothalamus. CONCLUSION: QFZS exerts its regulatory effects on fever by regulating the metabolism of lysophospholipids and arachidonic acid and the regulation of inflammation via transient receptor potential ion channels channels.


Asunto(s)
Ácido Araquidónico , Medicamentos Herbarios Chinos , Fiebre , Hipotálamo , Inflamación , Lisofosfolípidos , Ratas Sprague-Dawley , Animales , Ratas , Masculino , Fiebre/tratamiento farmacológico , Fiebre/metabolismo , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/farmacología , Ácido Araquidónico/metabolismo , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/genética , Humanos , Lisofosfolípidos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Hipertermia/tratamiento farmacológico , Hipertermia/metabolismo , Hipertermia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA