Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Appl Physiol (1985) ; 135(5): 1186-1198, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37795530

ABSTRACT

Epidemiological studies report higher risks of cardiovascular disease in humans exposed to heat stroke earlier in life. Previously, we explored mechanistic links between heat stroke and developing cardiac abnormalities using a preclinical mouse model of exertional heat stroke (EHS). Profound metabolic abnormalities developed in the ventricles of females but not males after 2 wk of recovery. Here we tested whether this lack of response in males could be attributed to the lower exercise performances or reduced thermal loads they experienced with the same running protocol. We systematically altered environmental temperature (Te) during EHS to manipulate heat exposure and exercise performance in the males. Three groups of adult C57BL/6 male mice were studied: "EHS-34" (Te = 34°C), "EHS-41" (Te = 41°C), and "EHS-39.5" (Te = 39.5°C). Mice ran until symptom limitation (unconsciousness), reaching max core temperature (Tc,max). After a 2-wk recovery, the mice were euthanized, and the ventricles were removed for untargeted metabolomics. Results were compared against age-matched nonexercise controls. The EHS-34 mice greatly elevated their exercise performance but reached lower Tc,max and lower thermal loads. The EHS-41 mice exhibited equivalent thermal loads, exercise times, and Tc,max compared with EHS-39.5. The ventricles from EHS-34 mice exhibited the greatest metabolic disturbances in the heart, characterized by shifts toward glucose metabolism, reductions in acylcarnitines, increased amino acid metabolites, elevations in antioxidants, altered TCA cycle flux, and increased xenobiotics. In conclusion, delayed metabolic disturbances following EHS in male myocardium appear to be greatly amplified by higher levels of exertion in the heat, even with lower thermal loads and max core temperatures.NEW & NOTEWORTHY Epidemiological data demonstrate greater cardiovascular risk in patients with previous heat stroke exposure. Using a preclinical mouse model of exertional heat stroke, male mice were exposed to one of three environmental temperatures (Te) during exercise. Paradoxically, after 2 wk, the mice in the lowest Te, exhibiting the largest exercise response and lowest heat load, had the greatest ventricular metabolic disturbances. Metabolic outcomes resemble developing left ventricular hypertrophy or stress-induced heart disease.


Subject(s)
Heat Stroke , Physical Exertion , Humans , Adult , Female , Male , Animals , Mice , Physical Exertion/physiology , Mice, Inbred C57BL , Body Temperature/physiology , Myocardium
2.
PLoS One ; 17(10): e0275715, 2022.
Article in English | MEDLINE | ID: mdl-36227921

ABSTRACT

INTRODUCTION: The capability of male mice to exercise in hot environments without succumbing to exertional heat stroke (EHS) is markedly blunted compared to females. Epidemiological evidence in humans and other mammals also suggests some degree of greater vulnerability to heat stroke in males compared to females. The origins of these differences are unknown, but testosterone has previously been shown to induce faster elevations in core temperature during acute, passive heat exposure. In this study, we tested the hypothesis that loss of testosterone and related sex hormones through castration would improve the performance and heat tolerance of male mice during EHS exposure. METHODS: Twenty-four male mice were randomly divided into 3 groups, untreated EHS mice (SHAM-EHS), castrated EHS mice (CAS+EHS) and naïve exercise controls (NAIVE). Exercise performance and physiological responses in the heat were monitored during EHS and early recovery. Two weeks later, blood and tissues were collected and analyzed for biomarkers of cardiac damage and testosterone. RESULTS: Core temperature in CAS+EHS rose faster to 39.5°C in the early stages of the EHS trial (P<0.0001). However, both EHS groups ran similar distances, exhibited similar peak core temperatures and achieved similar exercise times in the heat, prior to symptom limitation (unconsciousness). CAS+EHS mice had ~10.5% lower body mass at the time of EHS, but this provided no apparent advantage in performance. There was no evidence of myocardial damage in any group, and testosterone levels were undetectable in CAS+EHS after gonadectomy. CONCLUSIONS: The results of these experiments exclude the hypothesis that reduced performance of male mice during EHS trials is due to the effects of male sex hormones or intact gonads. However, the results are consistent with a role of male sex hormones or intact gonads in suppressing the early and rapid rise in core temperature during the early stages of exercise in the heat.


Subject(s)
Heat Stroke , Animals , Biomarkers , Female , Heat Stroke/diagnosis , Hot Temperature , Humans , Male , Mammals , Mice , Orchiectomy , Testosterone
3.
Shock ; 57(4): 600-607, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34798635

ABSTRACT

ABSTRACT: Skeletal muscles play important roles in innate immunity. However, in vitro, their sensitivity to LPS is low. In other tissues, LPS sensing is facilitated by the presence of plasma, LPS binding protein (LBP), or soluble CD14 (sCD14). This study addressed whether these are critical for LPS sensitivity in skeletal muscle and whether LPS responsiveness is different between slow versus fast muscle. Soleus (SOL) or extensor digitorum longus (EDL) muscles from adult male C57bl/6 mice were mounted in 1 mL oxygenated baths containing: buffer only; buffer+1% mouse plasma; buffer+1 µg/mL LBP; or buffer+1% plasma from sCD14-/- mice. In each condition, muscles were exposed to LPS from 0 µg/mL to 1.0 µg/mL. Bath samples were collected at 0, 1, and 2 h, and analyzed using cytokine multiplex arrays. In both SOL and EDL the predominant responding cytokines/chemokines were KC(CXCL1), IL-6, and MCP-1(CCL2) and their average responses were amplified by ∼10-fold in the presence of 1% plasma. Overall, SOL and EDL exhibited similar secretory responses in the presence of 1% plasma, with a lower limit of sensitivity to LPS of 0.01 µg/mL. LBP supplementation did not augment secretion; however, 1% plasma from CD14-/- mice suppressed cytokine/chemokine secretion from EDL muscle. In conclusion, intact slow and fast mouse muscles have similar cytokine/chemokine responses to LPS but depend on the presence of low levels of plasma constituents. Though sCD14 plays some role in EDL muscle, neither sCD14 nor LBP can fully account for the strong effects of plasma on LPS sensitivity.


Subject(s)
Lipopolysaccharide Receptors , Lipopolysaccharides , Animals , Cytokines/metabolism , Gene Expression , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Muscle, Skeletal/metabolism
4.
Sci Rep ; 11(1): 7316, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795743

ABSTRACT

Skeletal muscles secrete a wide variety of immunologically active cytokines, but the functional significance of this response to in vivo innate immunity is not understood. We addressed this by knocking out the toll receptor adapter protein, Myd88, only in skeletal muscle fibers (skmMyd88KO), and followed male and female mice at 6 and 12 h after peritoneal injection of cecal slurry (CS), a model of polymicrobial sepsis. Because of a previously identified increase in mortality to CS injection, males received ~ 30% lower dose. At 12 h, skmMyd88KO caused significant reductions in a wide variety of pro- and anti-inflammatory plasma cytokines, e.g. TNFα, IL-1ß and IL-10, compared to strain-matched controls in both males and females. Similar reductions were observed at 6 h in females. SkmMyd88KO led to ~ 40-50% elevations in peritoneal neutrophils at 6 and 12 h post CS in females. At 12 h post CS, skmMyd88KO increased peritoneal monocytes/macrophages and decreased %eosinophils and %basophils in females. SkmMyd88KO also led to significantly higher rates of mortality in female mice but not in males. In conclusion, the results suggest that skeletal muscle Myd88-dependent signal transduction can play functionally important role in normal whole body, innate immune inflammatory responses to peritoneal sepsis.


Subject(s)
Muscle, Skeletal/physiopathology , Myeloid Differentiation Factor 88/metabolism , Sepsis/microbiology , Sepsis/physiopathology , Signal Transduction , Animals , Cytokines , Disease Models, Animal , Female , Immune System , Immunity, Innate , Inflammation , Leukocytes/metabolism , Macrophages, Peritoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils , Peritoneum , Sepsis/immunology , Sex Factors , Time Factors
5.
J Physiol ; 599(1): 119-141, 2021 01.
Article in English | MEDLINE | ID: mdl-33037634

ABSTRACT

KEY POINTS: Exposure to exertional heat stroke (EHS) has been linked to increased long-term decrements of health. Epigenetic reprogramming is involved in the response to heat acclimation; however, whether the long-term effects of EHS are mediated by epigenetic reprogramming is unknown. In female mice, we observed DNA methylation reprogramming in bone marrow-derived (BMD) monocytes as early as 4 days of recovery from EHS and as late as 30 days compared with sham exercise controls. Whole blood, collected after 30 days of recovery from EHS, exhibited an immunosuppressive phenotype when challenged in vitro by lipopolysaccharide. After 30 days of recovery from EHS, BMD monocytes exhibited an altered in vitro heat shock response. The location of differentially methylated CpGs are predictive of both the immunosuppressive phenotype and altered heat shock responses. ABSTRACT: Exposure to exertional heat stroke (EHS) has been linked to increased susceptibility to a second heat stroke, infection and cardiovascular disease. Whether these clinical outcomes are mediated by an epigenetic memory is unknown. Using a preclinical mouse model of EHS, we investigated whether EHS exposure produces a lasting epigenetic memory in monocytes and whether there are phenotypic alterations that may be consistent with these epigenetic changes. Female mice underwent forced wheel running at 37.5°C/40% relative humidity until symptom limitation, characterized by CNS dysfunction. Results were compared with matched exercise controls at 22.5°C. Monocytes were isolated from bone marrow after 4 or 30 days of recovery to extract DNA and analyse methylation. Broad-ranging alterations to the DNA methylome were observed at both time points. At 30 days, very specific alterations were observed to the promoter regions of genes involved with immune responsiveness. To test whether these changes might be related to phenotype, whole blood at 30 days was challenged with lipopolysaccharide (LPS) to measure cytokine secretion; monocytes were also challenged with heat shock to quantify mRNA expression. Whole blood collected from EHS mice showed markedly attenuated inflammatory responses to LPS challenge. Furthermore, monocyte mRNA from EHS mice showed significantly altered responses to heat shock challenge. These results demonstrate that EHS leads to a unique DNA methylation pattern in monocytes and altered immune and heat shock responsiveness after 30 days. These data support the hypothesis that EHS exposure can induce long-term physiological changes that may be linked to altered epigenetic profiles.


Subject(s)
Heat Stroke , Motor Activity , Animals , Epigenesis, Genetic , Female , Heat Stroke/genetics , Heat-Shock Response/genetics , Immunosuppression Therapy , Mice
6.
Exp Physiol ; 106(1): 222-232, 2021 01.
Article in English | MEDLINE | ID: mdl-32281170

ABSTRACT

NEW FINDINGS: What is the central question of this study? Exertional heat stroke is accompanied by a marked inflammatory response. In this study, we explored the time course of acute phase proteins during recovery from severe heat stress in mice and the potential role of skeletal muscles as their source. What is the main finding and its importance? Exertional heat stroke transiently increased expression of acute phase proteins in mouse liver and plasma and depleted liver and plasma fibrinogen, a typical response to severe trauma. In contrast, skeletal muscle fibrinogen production was stimulated by heat stroke, which can provide an additional reservoir for fibrinogen supply to maintain the clotting potential throughout the body and locally within the muscle. ABSTRACT: Exertional heat stroke (EHS), the most severe manifestation of heat illness, is accompanied by a marked inflammatory response. The release of acute phase proteins (APPs) is an important component of inflammation, which can assist in tissue survival/repair. The time course of APPs in recovery from EHS is unknown. Furthermore, skeletal muscles produce APPs during infection, but it is unknown whether they can produce APPs after EHS. Our objective was to determine the time course of representative APPs in liver, plasma and skeletal muscle during recovery from EHS. Male C57BL6/J mice ran in a forced running wheel at 37.5°C, 40% relative humidity until symptom limitation. Exercise control (EXC) mice ran for the same duration and intensity at 22.5°C. Samples were collected (n = 6-12 per group) over 14 days of recovery. Protein abundance was quantified using immunoblots. Total and phosphorylated STAT3 (pSTAT3) at Tyr705, responsible for APP activation, increased in liver at 0.5 h after EHS compared with EXC, (P < 0.05 and P < 0.001, respectively). In contrast, in tibialis anterior (TA) muscle, total STAT3 increased at 3 h (P < 0.05) but pSTAT3 (Tyr705) did not. Liver serum amyloid A1 (SAA1) increased at 3 and 24 h after EHS (P < 0.05), whereas plasma SAA1 increased only at 3 h (P < 0.05). SAA1 was not detected in TA muscle. In liver and plasma, fibrinogen decreased at 3 h (P < 0.01) and increased in TA muscle (P < 0.05). Lipocalin-2 was undetectable in liver or TA muscle. Recovery from EHS is characterized by a transient acute phase response in both liver and skeletal muscle. However, APP expression profiles and subtypes differ between skeletal muscle and liver.


Subject(s)
Acute-Phase Reaction/physiopathology , Heat Stroke/physiopathology , Heat-Shock Response/physiology , Physical Exertion/physiology , Animals , Mice, Inbred C57BL , Muscle, Skeletal/physiopathology , Physical Conditioning, Animal/physiology
7.
Shock ; 55(5): 676-685, 2021 05 01.
Article in English | MEDLINE | ID: mdl-32826815

ABSTRACT

ABSTRACT: Interleukin-6 (IL-6) is a major cytokine released by skeletal muscle. Although IL-6 plays complex but well-known roles in host defense, the specific contribution of skeletal muscle IL-6 to innate immunity remains unknown. We tested its functional relevance by exposing inducible skeletal muscle IL-6 knockdown (skmIL-6KD) mice to a cecal slurry model of polymicrobial peritonitis and compared responses to strain-matched controls and skeletal muscle Cre-matched controls at 3, 6, and 12 h postinfection. In both sexes, skmIL-6KD mice at 6 h of infection exhibited marked changes to leukocyte trafficking in the peritoneum, characterized by ∼1.75-fold elevation in %neutrophils, a ∼3-fold reduction in %lymphocytes and a ∼2 to 3-fold reduction in %basophils. A similar pattern was seen at 12 h. No changes were observed in plasma leukocyte counts. Circulating cytokines in female skmIL-6KD mice at 6 h consistently showed modest reductions in IL-6, but marked reductions in a broad range of both pro- and anti-inflammatory cytokines, e.g., TNFα and IL-10. In both sexes at 12 h, a generalized suppression of plasma cytokines was also seen after the effects of Cre-induction with raloxifene were addressed. There were no significant effects of skmIL-6KD on mortality in either sex. Collectively, our results are consistent with skmIL-6 playing an important and previously unrecognized role in immune cell trafficking and cytokine regulation during septic shock.


Subject(s)
Immunity, Innate , Interleukin-6/physiology , Shock, Septic/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal
8.
Med Sci Sports Exerc ; 52(9): 1870-1878, 2020 09.
Article in English | MEDLINE | ID: mdl-32175974

ABSTRACT

Intestinal injury is one of the most prominent features of organ damage in exertional heat stroke (EHS). However, whether damage to the intestine in this setting is exacerbated by ibuprofen (IBU), the most commonly used nonsteroidal anti-inflammatory drug in exercising populations, is not well understood. PURPOSE: We hypothesized that IBU would exacerbate intestinal injury, reduce exercise performance, and increase susceptibility to heat stroke. METHODS: To test this hypothesis, we administered IBU via diet to male and female C57/BL6J mice, over 48 h before EHS. Susceptibility to EHS was determined by assessing exercise response using a forced running wheel, housed inside an environmental chamber at 37.5°C. Core temperature (Tc) was monitored by telemetry. Mice were allocated into four groups: exercise only (EXC); EHS + IBU; EXC + IBU; and EHS only. Exercise performance and Tc profiles were evaluated and stomachs, intestines and plasma were collected at 3 h post-EHS. RESULTS: The EHS + IBU males ran approximately 87% longer when Tc was above 41°C (P < 0.03) and attained significantly higher peak Tc (P < 0.01) than EHS-only mice. Histological analyses showed decreased villi surface area throughout the small intestine for both sexes in the EXC + IBU group versus EXC only. Interestingly, though EHS in both sexes caused intestinal injury, in neither sex were there any additional effects of IBU. CONCLUSIONS: Our results suggest that in a preclinical mouse model of EHS, oral IBU at pharmacologically effective doses does not pose additional risks of heat stroke, does not reduce exercise performance, and does not contribute further to intestinal injury, though this could have been masked by significant gut injury induced by EHS alone.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Heat Stroke/pathology , Ibuprofen/adverse effects , Intestines/pathology , Physical Conditioning, Animal/adverse effects , Animals , Biomarkers/blood , Dinoprostone/blood , Fatty Acid-Binding Proteins/blood , Female , Heat Stroke/etiology , Intestinal Mucosa/pathology , Intestines/drug effects , Leukocyte Count , Male , Mice, Inbred C57BL , Physical Conditioning, Animal/physiology , Physical Endurance/physiology , Running/physiology
9.
J Physiol ; 598(5): 967-985, 2020 03.
Article in English | MEDLINE | ID: mdl-32026469

ABSTRACT

KEY POINTS: Exposure to exertional heat stroke (EHS) is associated with increased risk of long-term cardiovascular disorders in humans. We demonstrate that in female mice, severe EHS results in metabolic changes in the myocardium, emerging only after 9-14 days. This was not observed in males that were symptom-limited at much lower exercise levels and heat loads compared to females. At 14 days of recovery in females, there were marked elevations in myocardial free fatty acids, ceramides and diacylglycerols, consistent with development of underlying cardiac abnormalities. Glycolysis shifted towards the pentose phosphate and glycerol-3-phosphate dehydrogenase pathways. There was evidence for oxidative stress, tissue injury and microscopic interstitial inflammation. The tricarboxylic acid cycle and nucleic acid metabolism pathways were also negatively affected. We conclude that exposure to EHS in female mice has the capacity to cause delayed metabolic disorders in the heart that could influence long-term health. ABSTRACT: Exposure to exertional heat stroke (EHS) is associated with a higher risk of long-term cardiovascular disease in humans. Whether this is a cause-and-effect relationship remains unknown. We studied the potential of EHS to contribute to the development of a 'silent' form of cardiovascular disease using a preclinical mouse model of EHS. Plasma and ventricular myocardial samples were collected over 14 days of recovery. Male and female C57bl/6J mice underwent forced wheel running for 1.5-3 h in a 37.5°C/40% relative humidity until symptom limitation, characterized by CNS dysfunction. They reached peak core temperatures of 42.2 ± 0.3°C. Females ran ∼40% longer, reaching ∼51% greater heat load. Myocardial and plasma samples (n = 8 per group) were obtained between 30 min and 14 days of recovery, analysed using metabolomics/lipidomics platforms and compared to exercise controls. The immediate recovery period revealed an acute energy substrate crisis from which both sexes recovered within 24 h. However, at 9-14 days, the myocardium of female mice developed marked elevations in free fatty acids, ceramides and diacylglycerols. Glycolytic and tricarboxylic acid cycle metabolites revealed bottlenecks in substrate flow, with build-up of intermediate metabolites consistent with oxidative stress and damage. Males exhibited only late stage reductions in acylcarnitines and elevations in acetylcarnitine. Histopathology at 14 days showed interstitial inflammation in the female hearts only. The results demonstrate that the myocardium of female mice is vulnerable to a slowly emerging metabolic disorder following EHS that may harbinger long-term cardiovascular complications. Lack of similar findings in males may reflect their lower heat exposure.


Subject(s)
Heat Stroke , Motor Activity , Animals , Female , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Myocardium
10.
J Appl Physiol (1985) ; 125(3): 841-849, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29901435

ABSTRACT

With increasing participation of females in endurance athletics and active military service, it is important to determine if there are inherent sex-dependent susceptibilities to exertional heat injury or heat stroke. In this study we compared responses of male and female adult mice to exertional heat stroke (EHS). All mice were instrumented for telemetry core temperature measurements and were exercise-trained for 3 wk before EHS. During EHS, environmental temperature was 37.5°C (35% RH) while the mice ran on a forced running wheel, using incremental increases in speed. The symptom-limited endpoint was loss of consciousness, occurring at ~42.2°C core temperature. Females ran greater distances (623 vs. 346 m, P < 0.0001), reached faster running speeds (7.2 vs. 5.1 m/min, P < 0.0001), exercised for longer times (177 vs. 124 min, P < 0.0001), and were exposed to greater internal heat loads (240 vs.160°C·min; P < 0.0001). Minimum Tc during hypothermic recovery was ~32.0°C in both sexes. Females lost 9.2% body weight vs. 7.5% in males ( P < 0.001). Females demonstrated higher circulating corticosterone (286 vs 183 ng/ml, P = 0.001, at 3 h), but most plasma cytokines were not different. A component of performance in females could be attributed to greater body surface area/mass and greater external power performance. However, there were significant and independent effects of sex alone and a crossed effect of "sex × power" on performance. These results demonstrate that female mice have greater resistance to EHS during exercise in hyperthermia and that these effects cannot be attributed solely to body size. NEW & NOTEWORTHY Female mice are surprisingly more resistant to exertional heat stroke than male mice. They run faster and longer and can withstand greater internal heat loads. These changes cannot be fully accounted for by increased body surface/mass ratio in females or on differences in aerobic performance. Although the stress-immune response in males and females was similar, females exhibited markedly higher plasma corticosteroid levels, which were sustained over 14 days of recovery.


Subject(s)
Heat Stroke/physiopathology , Animals , Body Size , Body Temperature , Corticosterone/blood , Cytokines/blood , Female , Male , Mice , Mice, Inbred C57BL , Physical Conditioning, Animal/physiology , Physical Endurance , Physical Exertion , Running/physiology , Sex Characteristics , Weight Loss
11.
Shock ; 50(2): 226-232, 2018 08.
Article in English | MEDLINE | ID: mdl-28957876

ABSTRACT

Sepsis continues to be a major challenge for modern medicine. Several preclinical models were developed to study sepsis and each has strengths and weaknesses. The cecal slurry (CS) method is a practical alternative because it does not require surgery, and the infection can be dosed. However, one disadvantage is that the dosage must be determined for each CS preparation using survival studies. Our aim was to refine a survival protocol for the CS model by determining a premonitory humane endpoint that would reduce animal suffering. Mice become hypothermic in sepsis; therefore, we tested whether reductions in surface temperature (Ts), measured by noninvasive infrared thermometry, could predict eventual death. We injected 154 C57BL/6J mice with CS (0.9-1.8 mg/g) and periodically monitored Ts at the xiphoid process over 5 days. We used, as predictors, combinations of temperature thresholds (29°C -31°C) and times, postinjection (18-36 h). A receiver-operator curve, sensitivity, and specificity were determined. A Distress Index value was calculated for the threshold conditions. The optimum detection threshold (highest Youden index) was found at Ts ≤ 30.5°C at 24 h (90% specific, 84% sensitive). This threshold condition reduced animal suffering by 41% while providing an accurate survival rate estimate. Using this threshold, only 13 of 154 mice would have died from sepsis; 67 would have been euthanized at 24 h, and only 7 of 154 would have been euthanized unnecessarily. In conclusion, using a humane endpoint of Ts ≤ 30.5°C at 24 h accurately predicts mortality and can effectively reduce animal suffering during CS survival protocols.


Subject(s)
Body Temperature , Hypothermia , Shock, Septic/physiopathology , Xiphoid Bone/physiopathology , Animals , Disease Models, Animal , Female , Male , Mice , Predictive Value of Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...