Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Life Sci ; 346: 122633, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38615746

ABSTRACT

AIMS: Systemic administration of ammonium chloride (NH4Cl), an acidifying agent used in human patients and experimental conditions, causes hypothermia in mice, however, the mechanisms of the thermoregulatory response to NH4Cl and whether it develops in other species remained unknown. MAIN METHODS: We studied body temperature (Tb) changes in rats and mice induced by intraperitoneal administration of NH4Cl after blockade of transient receptor potential vanilloid-1 (TRPV1) or ankyrin-1 (TRPA1) channels. KEY FINDINGS: In rats, NH4Cl decreased Tb by 0.4-0.8°C (p < 0.05). The NH4Cl-induced hypothermia also developed in Trpv1 knockout (Trpv1-/-) and wild-type (Trpv1+/+) mice, however, the Tb drop was exaggerated in Trpv1-/- mice compared to Trpv1+/+ controls with maximal decreases of 4.0 vs. 2.1°C, respectively (p < 0.05). Pharmacological blockade of TRPV1 channels with AMG 517 augmented the hypothermic response to NH4Cl in genetically unmodified mice and rats (p < 0.05 for both). In contrast, when NH4Cl was infused to mice genetically lacking the TRPA1 channel, the hypothermic response was significantly attenuated compared to wild-type controls with maximal mean Tb difference of 1.0°C between the genotypes (p = 0.008). Pretreatment of rats with a TRPA1 antagonist (A967079) also attenuated the NH4Cl-induced Tb drop with a maximal difference of 0.7°C between the pretreatment groups (p = 0.003). SIGNIFICANCE: TRPV1 channels limit, whereas TRPA1 channels exaggerate the development of NH4Cl-induced hypothermia in rats and mice, but other mechanisms are also involved. Our results warrant for regular Tb control and careful consideration of NH4Cl treatment in patients with TRPA1 and TRPV1 channel dysfunctions.


Subject(s)
Hypothermia , TRPA1 Cation Channel , TRPV Cation Channels , Animals , Male , Mice , Rats , Ammonium Chloride/pharmacology , Body Temperature/drug effects , Hypothermia/chemically induced , Hypothermia/metabolism , Mice, Inbred C57BL , Mice, Knockout , Rats, Sprague-Dawley , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
2.
Temperature (Austin) ; 10(1): 136-154, 2023.
Article in English | MEDLINE | ID: mdl-37187834

ABSTRACT

We identified the neural pathway of the hyperthermic response to TRPV1 antagonists. We showed that hyperthermia induced by i.v. AMG0347, AMG 517, or AMG8163 did not occur in rats with abdominal sensory nerves desensitized by pretreatment with a low i.p. dose of resiniferatoxin (RTX, TRPV1 agonist). However, neither bilateral vagotomy nor bilateral transection of the greater splanchnic nerve attenuated AMG0347-induced hyperthermia. Yet, this hyperthermia was attenuated by bilateral high cervical transection of the spinal dorsolateral funiculus (DLF). To explain the extra-splanchnic, spinal mediation of TRPV1 antagonist-induced hyperthermia, we proposed that abdominal signals that drive this hyperthermia originate in skeletal muscles - not viscera. If so, in order to prevent TRPV1 antagonist-induced hyperthermia, the desensitization caused by i.p. RTX should spread into the abdominal-wall muscles. Indeed, we found that the local hypoperfusion response to capsaicin (TRPV1 agonist) in the abdominal-wall muscles was absent in i.p. RTX-desensitized rats. We then showed that the most upstream (lateral parabrachial, LPB) and the most downstream (rostral raphe pallidus) nuclei of the intrabrain pathway that controls autonomic cold defenses are also required for the hyperthermic response to i.v. AMG0347. Injection of muscimol (inhibitor of neuronal activity) into the LPB or injection of glycine (inhibitory neurotransmitter) into the raphe blocked the hyperthermic response to i.v. AMG0347, whereas i.v. AMG0347 increased the number of c-Fos cells in the raphe. We conclude that the neural pathway of TRPV1 antagonist-induced hyperthermia involves TRPV1-expressing sensory nerves in trunk muscles, the DLF, and the same LPB-raphe pathway that controls autonomic cold defenses.

3.
Sci Rep ; 13(1): 588, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36631486

ABSTRACT

Macrophage migration inhibitory factor (MIF) has been considered as a biomarker in sepsis, however the predictive value of the pattern of its kinetics in the serum and in the urine has remained unclarified. It is also unclear whether the kinetics of MIF are different between males and females. We conducted a single-center prospective, observational study with repeated measurements of MIF in serum and urine on days 0, 2, and 4 from admission to the intensive care unit (ICU) in 50 adult septic patients. We found that in patients who died within 90 days, there was an increase in serum MIF level from day 0 to 4, whereas in the survivors there was rather a decrease (p = 0.018). The kinetics were sex-dependent as the same difference in the pattern was present in males (p = 0.014), but not in females (p = 0.418). We also found that urine MIF was markedly lower in patients who died than in survivors of sepsis (p < 0.050). Urine MIF levels did not show temporal changes: there was no meaningful difference between day 0 and 4. These results suggest that kinetics of serum MIF during the initial days from ICU admission can predict death, especially in male patients. Additionally, lower urine MIF levels can also indicate death without showing meaningful temporal kinetics.


Subject(s)
Macrophage Migration-Inhibitory Factors , Sepsis , Adult , Female , Humans , Male , Biomarkers , Intensive Care Units , Macrophage Migration-Inhibitory Factors/blood , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/urine , Prospective Studies , Sepsis/complications , Sepsis/diagnosis
4.
Eur J Med Chem ; 247: 115050, 2023 Feb 05.
Article in English | MEDLINE | ID: mdl-36587420

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with enzymatic activities. Anti-inflammatory effects of MIF enzyme inhibitors indicate a link between its cytokine- and catalytic activities. Herein the synthesis, docking, and bioactivity of substituted benzylidene-1-indanone and -1-tetralone derivatives as MIF-tautomerase inhibitors is reported. Many of these substituted benzylidene-1-tetralones and -indan-1-ones were potent MIF-tautomerase inhibitors (IC50 < 10 µmol/L), and the most potent inhibitors were the 1-indanone derivatives 16 and 20. Some of these compounds acted as selective enolase or ketonase inhibitors. In addition, compounds 16, 20, 26, 37 and 61 efficiently inhibited NO, TNFα and IL-6 production in lipopolysaccharide-induced macrophages. Compound 20, 37 and 61 also inhibited ROS generation, and compound 26 and 37 abolished activation of NF-κB. Compound 37 significantly augmented hypothermia induced by high dose of lipopolysaccharide in mice. The possible mechanisms of action were explored using molecular modelling and docking, as well as molecular dynamics simulations.


Subject(s)
Macrophage Migration-Inhibitory Factors , Shock, Septic , Animals , Mice , Lipopolysaccharides/pharmacology , Shock, Septic/chemically induced , Shock, Septic/drug therapy , Molecular Dynamics Simulation
5.
BMJ Open ; 12(10): e059271, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36261235

ABSTRACT

OBJECTIVES: Heart rate (HR) is one of the physiological variables in the early assessment of trauma-related haemorrhagic shock, according to Advanced Trauma Life Support (ATLS). However, its efficiency as predictor of mortality is contradicted by several studies. Furthermore, the linear association between HR and the severity of shock and blood loss presented by ATLS is doubtful. This systematic review aims to update current knowledge on the role of HR in the initial haemodynamic assessment of patients who had a trauma. DESIGN: This study is a systematic review and meta-regression that follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses recommendations. DATA SOURCES: EMBASE, MEDLINE, CENTRAL and Web of Science databases were systematically searched through on 1 September 2020. ELIGIBILITY CRITERIA: Papers providing early HR and mortality data on bleeding patients who had a trauma were included. Patient cohorts were considered haemorrhagic if the inclusion criteria of the studies contained transfusion and/or positive focused assessment with sonography for trauma and/or postinjury haemodynamical instability and/or abdominal gunshot injury. Studies on burns, traumatic spinal or brain injuries were excluded. Papers published before January 2010 were not considered. DATA EXTRACTION AND SYNTHESIS: Data extraction and risk of bias were assessed by two independent investigators. The association between HR and mortality of patients who had a trauma was assessed using meta-regression analysis. As subgroup analysis, meta-regression was performed on patients who received blood products. RESULTS: From a total of 2017 papers, 19 studies met our eligibility criteria. Our primary meta-regression did not find a significant relation (p=0.847) between HR and mortality in patients who had a trauma with haemorrhage. Our subgroup analysis included 10 studies, and it could not reveal a linear association between HR and mortality rate. CONCLUSIONS: In accordance with the literature demonstrating the multiphasic response of HR to bleeding, our study presents the lack of linear association between postinjury HR and mortality. Modifying the pattern of HR derangements in the ATLS shock classification may result in a more precise teaching tool for young clinicians.


Subject(s)
Shock, Hemorrhagic , Humans , Shock, Hemorrhagic/etiology , Advanced Trauma Life Support Care , Tachycardia , Hemorrhage/etiology , Heart Rate
6.
Injury ; 53(12): 3912-3919, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36216616

ABSTRACT

BACKGROUND: There are several options available for conservative treatment of partial-thickness burns, however, reliable, affordable, and easily obtainable animal testing models are hard to find for the comparison of the different treatment methods. We aimed at developing a preclinical testing model and at comparing four treatment methods for superficial partial-thickness burns. METHODS: Burn injury was induced in 90 adult male Wistar rats by placing the 130°C hot tip of a commercially obtainable soldering device for 30 s on the clipped skin of the interscapular region at a steady pressure. Skin histology was studied on days 5, 10, and 22 after the induction of the burn injury, on which days, respectively, the ratio of the not epithelialized wound (%), the extent of re-epithelialization (score), and the scar thickness (µm) were assessed. We compared 4 groups: silver-sulfadiazine cream, zinc-hyaluronan gel, silver foam dressing, and the combination of zinc-hyaluronan gel with a silver foam dressing. RESULTS: On day 5, the induction of superficial partial-thickness burn injury was confirmed histologically in the rats. The zinc-hyaluronan gel and the combination treatment resulted in a markedly smaller ratio of the non-epithelialized area (29 ± 10% and 28 ± 13%, respectively) than silver-sulfadiazine cream (69 ± 4%; p < 0.01). On day 10, the extent of re-epithelialization was the lowest (∼0.2) in the silver-sulfadiazine cream group, while the other 3 treatments performed significantly better. The combination treatment lead to the maximal score of 2 in all rats, which was higher than in the other 3 treatment groups. On day 22, the scar thickness was the smallest in the combination treatment group (560 ± 42 µm), which was significantly less than in the silver-sulfadiazine cream group (712 ± 38 µm; p < 0.05). CONCLUSIONS: We designed and histologically confirmed a reproducible method for induction of superficial partial-thickness burns in rats for preclinical testing. In our model, the combination of zinc-hyaluronan gel with silver foam dressing was more effective than either of its components alone or than silver-sulfadiazine cream.


Subject(s)
Anti-Infective Agents, Local , Burns , Silver , Soft Tissue Injuries , Zinc , Animals , Male , Rats , Anti-Infective Agents, Local/therapeutic use , Bandages , Burns/drug therapy , Cicatrix/drug therapy , Cicatrix/pathology , Hyaluronic Acid/therapeutic use , Rats, Wistar , Silver/therapeutic use , Silver Sulfadiazine/therapeutic use , Soft Tissue Injuries/drug therapy , Zinc/therapeutic use
7.
Polymers (Basel) ; 14(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35160461

ABSTRACT

We report the additive manufacturing of a heat-exchange device that can be used as a cooling accessory in a wire myograph. Wire myography is used for measuring vasomotor responses in small resistance arteries; however, the commercially available devices are not capable of active cooling. Here, we critically evaluated a transparent resin material, in terms of mechanical, structural, and thermal behavior. Tensile strength tests (67.66 ± 1.31 MPa), Charpy impact strength test (20.70 ± 2.30 kJ/m2), and Shore D hardness measurements (83.0 ± 0.47) underlined the mechanical stability of the material, supported by digital microscopy, which revealed a glass-like structure. Differential scanning calorimetry with thermogravimetry analysis and thermal conductivity measurements showed heat stability until ~250 °C and effective heat insulation. The 3D-printed heat exchanger was tested in thermophysiology experiments measuring the vasomotor responses of rat tail arteries at different temperatures (13, 16, and 36 °C). The heat-exchange device was successfully used as an accessory of the wire myograph system to cool down the experimental chambers and steadily maintain the targeted temperatures. We observed temperature-dependent differences in the vasoconstriction induced by phenylephrine and KCl. In conclusion, the transparent resin material can be used in additive manufacturing of heat-exchange devices for biomedical research, such as wire myography. Our animal experiments underline the importance of temperature-dependent physiological mechanisms, which should be further studied to understand the background of the thermal changes and their consequences.

8.
Crit Rev Food Sci Nutr ; 62(25): 7025-7035, 2022.
Article in English | MEDLINE | ID: mdl-33840333

ABSTRACT

Abnormal cholesterol level is a major risk factor in the development of atherosclerosis, which is a fundamental derangement in cardiovascular diseases. Any efforts should be undertaken to lower blood cholesterol levels. Among dietary interventions, capsaicinoid supplementation is also considered as a novel cholesterol-lowering approach, but human studies concluded contradictory results about its effectiveness. The present meta-analysis aimed at determining the effects of capsaicinoids on serum lipid profile in humans. We searched the PubMed, EMBASE, and CENTRAL databases from inception to February 2021. We included 10 controlled studies, which involved 398 participants. We found that dietary capsaicinoid supplementation alone or in combination with other substances significantly (p = 0.004 and 0.001, respectively) reduced serum total cholesterol level compared to controls with an overall standardized mean difference of -0.52 (95% confidence interval: -0.83, -0.21). Capsaicinoids also decreased low-density lipoprotein level significantly (p = 0.035), whereas no effect was observed on serum levels of high-density lipoprotein and triglycerides. Our findings provide novel quantitative evidence for the efficacy of dietary capsaicin supplementation in lowering serum total cholesterol and low-density lipoprotein levels in humans. To validate our conclusion, further randomized controlled trials in a diverse population of adult humans receiving dietary capsaicinoid supplementation are warranted.


Subject(s)
Transient Receptor Potential Channels , Adult , Cholesterol , Cholesterol, HDL , Cholesterol, LDL , Dietary Supplements , Humans , Triglycerides
9.
Am J Physiol Endocrinol Metab ; 322(1): E10-E23, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34779255

ABSTRACT

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.


Subject(s)
Body Temperature/drug effects , Cholecystokinin/administration & dosage , Cyclooxygenase 2/metabolism , Hyperthermia/chemically induced , Hyperthermia/metabolism , Signal Transduction/drug effects , Animals , Anorexia/chemically induced , Benzodiazepines/administration & dosage , Body Temperature Regulation/drug effects , Cholecystokinin/adverse effects , Cyclooxygenase 2 Inhibitors/administration & dosage , Disease Models, Animal , Eating/drug effects , Fever/chemically induced , Fever/drug therapy , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Lipopolysaccharides/adverse effects , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Receptor, Cholecystokinin B/antagonists & inhibitors , Treatment Outcome
10.
Pharmaceuticals (Basel) ; 14(10)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34681216

ABSTRACT

Hydrogen sulfide (H2S) has been shown in previous studies to cause hypothermia and hypometabolism in mice, and its thermoregulatory effects were subsequently investigated. However, the molecular target through which H2S triggers its effects on deep body temperature has remained unknown. We investigated the thermoregulatory response to fast-(Na2S) and slow-releasing (GYY4137) H2S donors in C57BL/6 mice, and then tested whether their effects depend on the transient receptor potential ankyrin-1 (TRPA1) channel in Trpa1 knockout (Trpa1-/-) and wild-type (Trpa1+/+) mice. Intracerebroventricular administration of Na2S (0.5-1 mg/kg) caused hypothermia in C57BL/6 mice, which was mediated by cutaneous vasodilation and decreased thermogenesis. In contrast, intraperitoneal administration of Na2S (5 mg/kg) did not cause any thermoregulatory effect. Central administration of GYY4137 (3 mg/kg) also caused hypothermia and hypometabolism. The hypothermic response to both H2S donors was significantly (p < 0.001) attenuated in Trpa1-/- mice compared to their Trpa1+/+ littermates. Trpa1 mRNA transcripts could be detected with RNAscope in hypothalamic and other brain neurons within the autonomic thermoeffector pathways. In conclusion, slow- and fast-releasing H2S donors induce hypothermia through hypometabolism and cutaneous vasodilation in mice that is mediated by TRPA1 channels located in the brain, presumably in hypothalamic neurons within the autonomic thermoeffector pathways.

11.
J Neurotrauma ; 38(19): 2772-2776, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34002636

ABSTRACT

The benefits of therapeutic hypothermia (TH) in severe traumatic brain injury (sTBI) have been long debated. In 2018, the POLAR study, a high-quality international trial, appeared to end the debate by showing that TH did not improve mortality in sTBI. However, the POLAR-based recommendation to abandon TH was challenged by different investigators. In our recent meta-analysis, we introduced the cooling index (COIN) to assess the extent of cooling and showed that TH is beneficial in sTBI, but only when the COIN is sufficiently high. In the present study, we calculated the COIN for the POLAR study and ran a new meta-analysis, which included the POLAR data and accounted for the cooling extent. The POLAR study targeted a high cooling extent (COIN of 276°C × h; calculated for 72 h), but the achieved cooling was much lower (COIN of 193°C × h)-because of deviations from the protocol. When the POLAR data were included in the COIN-based meta-analysis, TH had an overall effect of reducing death (odds rate of 0.686; p = 0.007). Among the subgroups with different COIN levels, the only significantly decreased odds rate (i.e., beneficial effect of TH) was observed in the subgroup with high COIN (0.470; p = 0.013). We conclude that, because of deviations from the targeted cooling protocol, the overall cooling extent was not sufficiently high in the POLAR study, thus masking the beneficial effects of TH. The current analysis shows that TH is beneficial in sTBI, but only when the COIN is high. Abandoning the use of TH in sTBI may be premature.


Subject(s)
Brain Injuries, Traumatic/mortality , Brain Injuries, Traumatic/therapy , Hypothermia, Induced , Humans , Outcome Assessment, Health Care , Patient Selection
12.
Sci Rep ; 11(1): 8051, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33850259

ABSTRACT

The hunt for useful sepsis biomarkers is ongoing. Macrophage migration inhibitory factor (MIF) was implicated as a biomarker in sepsis, but its diagnostic and prognostic value has remained unclear in human studies. Here, we aimed at clarifying the value of MIF as a sepsis biomarker with the meta-analysis of clinical trials. PubMed, EMBASE, and Cochrane Central Register of Controlled Trials databases were searched until December 2019. From the included studies, blood MIF levels and indicators of disease severity were extracted in septic and control patient groups. Twenty-one eligible studies were identified, including data from 1876 subjects (of which 1206 had sepsis). In the septic patients, blood MIF levels were significantly higher than in healthy controls with a standardized mean difference (SMD) of 1.47 (95% confidence interval, CI: 0.96-1.97; p < 0.001) and also higher than in patient groups with nonseptic systemic inflammation (SMD = 0.94; CI: 0.51-1.38; p < 0.001). Markedly greater elevation in blood MIF level was found in the more severe forms of sepsis and in nonsurvivors than in less severe forms and in survivors with SMDs of 0.84 (CI: 0.45-1.24) and 0.75 (CI: 0.40-1.11), respectively (p < 0.001 for both). In conclusion, blood MIF level is more elevated in systemic inflammation caused by infection (i.e., sepsis) compared to noninfectious causes. In more severe forms of sepsis, including fatal outcome, MIF levels are higher than in less severe forms. These results suggest that MIF can be a valuable diagnostic and prognostic biomarker in sepsis given that well-designed clinical trials validate our findings.


Subject(s)
Macrophage Migration-Inhibitory Factors , Sepsis , Biomarkers/blood , Humans , Prognosis , Sepsis/blood
13.
PLoS One ; 14(9): e0223063, 2019.
Article in English | MEDLINE | ID: mdl-31553768

ABSTRACT

In pediatric burns the use of systemic antibiotic prophylaxis is a standard procedure in some burn centers, though its beneficial effect on the infectious complications is debated. The present meta-analysis aimed at determining whether systemic antibiotic prophylaxis prevents infectious complications in pediatric patients with burn injuries. We searched the PubMed, EMBASE, and Cochrane Library databases from inception to August 2019. We included 6 studies, in which event rates of infectious complications were reported in children with burn injuries receiving or not receiving systemic antibiotic prophylaxis. We found that the overall odds ratio (OR) of developing an infection (including local and systemic) was not different between the groups (OR = 1.35; 95% CI, 0.44, 4.18). The chances for systemic infectious complications alone were also not different between antibiotic-treated and non-treated patients (OR = 0.74; 95% CI, 0.38, 1.45). Based on the age, affected total body surface area, and country income level, we did not find any subgroup that benefited from the prophylaxis. Our findings provide quantitative evidence for the inefficacy of systemic antibiotic prophylaxis in preventing infections in pediatric burns. To validate our conclusion, multinational, randomized trials in a diverse population of children with burn injuries are warranted.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antibiotic Prophylaxis/methods , Bacterial Infections/epidemiology , Burns/complications , Antibiotic Prophylaxis/statistics & numerical data , Bacterial Infections/etiology , Bacterial Infections/prevention & control , Child , Humans , Incidence , Treatment Outcome
15.
Front Immunol ; 9: 166, 2018.
Article in English | MEDLINE | ID: mdl-29459872

ABSTRACT

Neurokinin (NK) signaling is involved in various inflammatory processes. A common manifestation of systemic inflammation is fever, which is usually induced in animal models with the administration of bacterial lipopolysaccharide (LPS). A role for the NK1 receptor was shown in LPS-induced fever, but the underlying mechanisms of how the NK1 receptor contributes to febrile response, especially in the early phase, have remained unknown. We administered LPS (120 µg/kg, intraperitoneally) to mice with the Tacr1 gene, i.e., the gene encoding the NK1 receptor, either present (Tacr1+/+ ) or absent (Tacr1-/- ) and measured their thermoregulatory responses, serum cytokine levels, tissue cyclooxygenase-2 (COX-2) expression, and prostaglandin (PG) E2 concentration. We found that the LPS-induced febrile response was attenuated in Tacr1-/- compared to their Tacr1+/+ littermates starting from 40 min postinfusion. The febrigenic effect of intracerebroventricularly administered PGE2 was not suppressed in the Tacr1-/- mice. Serum concentration of pyrogenic cytokines did not differ between Tacr1-/- and Tacr1+/+ at 40 min post-LPS infusion. Administration of LPS resulted in amplification of COX-2 mRNA expression in the lungs, liver, and brain of the mice, which was statistically indistinguishable between the genotypes. In contrast, the LPS-induced augmentation of COX-2 protein expression was attenuated in the lungs and tended to be suppressed in the liver of Tacr1-/- mice compared with Tacr1+/+ mice. The Tacr1+/+ mice responded to LPS with a significant surge of PGE2 production in the lungs, whereas Tacr1-/- mice did not. In conclusion, the NK1 receptor is necessary for normal fever genesis. Our results suggest that the NK1 receptor contributes to the early phase of LPS-induced fever by enhancing COX-2 protein expression in the periphery. These findings advance the understanding of the crosstalk between NK signaling and the "cytokine-COX-2-prostaglandin E2" axis in systemic inflammation, thereby open up the possibilities for new therapeutic approaches.


Subject(s)
Cyclooxygenase 2/metabolism , Fever/immunology , Lipopolysaccharides/adverse effects , Receptors, Neurokinin-1/metabolism , Animals , Cyclooxygenase 2/genetics , Cytokines/blood , Dinoprostone/blood , Female , Fever/chemically induced , Inflammation/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Neurokinin-1/genetics , Signal Transduction
16.
Anesthesiology ; 127(5): 813-823, 2017 11.
Article in English | MEDLINE | ID: mdl-28806222

ABSTRACT

BACKGROUND: Intraoperative hypothermia and postoperative pain control are two important clinical challenges in anesthesiology. Transient receptor potential vanilloid 1 has been implicated both in thermoregulation and pain. Transient receptor potential vanilloid 1 antagonists were not advanced as analgesics in humans in part due to a side effect of hyperthermia. This study tested the hypothesis that a single, preincision injection of a transient receptor potential vanilloid 1 antagonist could prevent anesthesia-induced hypothermia and decrease the opioid requirement for postsurgical hypersensitivity. METHODS: General anesthesia was induced in rats and mice with either isoflurane or ketamine, and animals were treated with transient receptor potential vanilloid 1 antagonists (AMG 517 or ABT-102). The core body temperature and oxygen consumption were monitored during anesthesia and the postanesthesia period. The effect of preincision AMG 517 on morphine-induced reversal of postincision hyperalgesia was evaluated in rats. RESULTS: AMG 517 and ABT-102 dose-dependently prevented general anesthesia-induced hypothermia (mean ± SD; from 1.5° ± 0.1°C to 0.1° ± 0.1°C decrease; P < 0.001) without causing hyperthermia in the postanesthesia phase. Isoflurane-induced hypothermia was prevented by AMG 517 in wild-type but not in transient receptor potential vanilloid 1 knockout mice (n = 7 to 11 per group). The prevention of anesthesia-induced hypothermia by AMG 517 involved activation of brown fat thermogenesis with a possible contribution from changes in vasomotor tone. A single preincision dose of AMG 517 decreased the morphine dose requirement for the reduction of postincision thermal (12.6 ± 3.0 vs. 15.6 ± 1.0 s) and mechanical (6.8 ± 3.0 vs. 9.5 ± 3.0 g) withdrawal latencies. CONCLUSIONS: These studies demonstrate that transient receptor potential vanilloid 1 antagonists prevent anesthesia-induced hypothermia and decrease opioid dose requirements for the reduction of postincisional hypersensitivity in rodents.


Subject(s)
Analgesics, Opioid/therapeutic use , Anesthesia/adverse effects , Hypothermia/chemically induced , Hypothermia/prevention & control , Pain, Postoperative/drug therapy , TRPV Cation Channels/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Animals , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Dose-Response Relationship, Drug , Female , Hypothermia/pathology , Indazoles/pharmacology , Indazoles/therapeutic use , Male , Mice , Mice, Knockout , Pain Measurement/drug effects , Pain Measurement/methods , Pain, Postoperative/pathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Urea/analogs & derivatives , Urea/pharmacology , Urea/therapeutic use
17.
J Vasc Res ; 53(5-6): 279-290, 2016.
Article in English | MEDLINE | ID: mdl-27923234

ABSTRACT

pH changes can influence local blood flow, but the mechanisms of how acids and bases affect vascular tone is not fully clarified. Transient receptor potential vanilloid-1 (TRPV1) channels are expressed in vessels and can be activated by pH alterations. Thus, we hypothesized that TRPV1 channels are involved in the mediation of vascular responses to acid-base changes. Vasomotor responses to HCl, NaOH, and capsaicin were measured in isolated murine carotid and tail skin arteries. The function of TRPV1 was blocked by either of three approaches: Trpv1 gene disruption, pharmacological blockade with a TRPV1 antagonist (BCTC), and functional impairment of mainly neural TRPV1 channels (desensitization). In each artery type of control mice, HCl caused relaxation but NaOH contraction, and both responses were augmented after genetic or pharmacological TRPV1 blockade. In arteries of TRPV1-desensitized mice, HCl-induced relaxation did not differ from controls, whereas NaOH-induced contraction was augmented. All three types of TRPV1 blockade had more pronounced effects in carotid than in tail skin arteries. We conclude that TRPV1 channels limit the vasomotor responses to changes in pH. While base-induced arterial contraction is regulated primarily by neural TRPV1 channels, acid-induced arterial relaxation is modulated by TRPV1 channels located on nonneural vascular structures.


Subject(s)
Acid-Base Equilibrium , Carotid Arteries/metabolism , Skin/blood supply , TRPV Cation Channels/metabolism , Vasoconstriction , Vasodilation , Vasomotor System/metabolism , Acid-Base Equilibrium/drug effects , Animals , Capsaicin/pharmacology , Carotid Arteries/drug effects , Carotid Arteries/innervation , Dose-Response Relationship, Drug , Female , Hydrochloric Acid/pharmacology , Hydrogen-Ion Concentration , In Vitro Techniques , Male , Mice, Inbred C57BL , Mice, Knockout , Rats, Wistar , Sodium Hydroxide/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/deficiency , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics , Tail , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Vasomotor System/drug effects
18.
Cell Cycle ; 14(8): 1260-7, 2015.
Article in English | MEDLINE | ID: mdl-25774749

ABSTRACT

Systemic inflammation is accompanied by an increased production of reactive oxygen species (ROS) and by either fever or hypothermia (or both). To study aseptic systemic inflammation, it is often induced in rats by the intravenous administration of bacterial lipopolysaccharide (LPS). Knowing that bilirubin is a potent ROS scavenger, we compared responses to LPS between normobilirubinemic Gunn rats (heterozygous, asymptomatic; J/+) and hyperbilirubinemic Gunn rats (homozygous, jaundiced; J/J) to establish whether ROS mediate fever and hypothermia in aseptic systemic inflammation. These two genotypes correspond to undisturbed versus drastically suppressed (by bilirubin) tissue accumulation of ROS, respectively. A low dose of LPS (10 µg/kg) caused a typical triphasic fever in both genotypes, without any intergenotype differences. A high dose of LPS (1,000 µg/kg) caused a complex response consisting of early hypothermia followed by late fever. The hypothermic response was markedly exaggerated, whereas the subsequent fever response was strongly attenuated in J/J rats, as compared to J/+ rats. J/J rats also tended to respond to 1,000 µg/kg with blunted surges in plasma levels of all hepatic enzymes studied (alanine aminotransferase, aspartate aminotransferase, gamma-glutamyl transferase), thus suggesting an attenuation of hepatic damage. We propose that the reported exaggeration of LPS-induced hypothermia in J/J rats occurs via direct inhibition of nonshivering thermogenesis by bilirubin and possibly via a direct vasodilatatory action of bilirubin in the skin. This hypothermia-exaggerating effect might be responsible, at least in part, for the observed tendency of J/J rats to be protected from LPS-induced hepatic damage. The attenuation of the fever response to 1,000 µg/kg could be due to either direct actions of bilirubin on thermoeffectors or the ROS-scavenging action of bilirubin. However, the experiments with 10 µg/kg strongly suggest that ROS signaling is not involved in the fever response to low doses of LPS.


Subject(s)
Hyperbilirubinemia/pathology , Hypothermia, Induced , Lipopolysaccharides/toxicity , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bilirubin/blood , Blood Urea Nitrogen , Creatinine/blood , Free Radical Scavengers/chemistry , Free Radical Scavengers/metabolism , Hyperbilirubinemia/metabolism , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Rats , Rats, Gunn , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism , gamma-Glutamyltransferase/blood
19.
J Mol Neurosci ; 54(3): 543-54, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24994541

ABSTRACT

Administration of the long form (38 amino acids) of pituitary adenylate cyclase-activating polypeptide (PACAP38) into the central nervous system causes hyperthermia, suggesting that PACAP38 plays a role in the regulation of deep body temperature (T b). In this study, we investigated the thermoregulatory role of PACAP38 in details. First, we infused PACAP38 intracerebroventricularly to rats and measured their T b and autonomic thermoeffector responses. We found that central PACAP38 infusion caused dose-dependent hyperthermia, which was brought about by increased thermogenesis and tail skin vasoconstriction. Compared to intracerebroventricular administration, systemic (intravenous) infusion of the same dose of PACAP38 caused significantly smaller hyperthermia, indicating a central site of action. We then investigated the thermoregulatory phenotype of mice lacking the Pacap gene (Pacap (-/-)). Freely moving Pacap (-/-) mice had higher locomotor activity throughout the day and elevated deep T b during the light phase. When the Pacap (-/-) mice were loosely restrained, their metabolic rate and T b were lower compared to their wild-type littermates. We conclude that PACAP38 causes hyperthermia via activation of the autonomic cold-defense thermoeffectors through central targets. Pacap (-/-) mice express hyperkinesis, which is presumably a compensatory mechanism, because under restrained conditions, these mice are hypometabolic and hypothermic compared to controls.


Subject(s)
Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Thermogenesis/drug effects , Animals , Female , Fever/genetics , Fever/metabolism , Male , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Rats , Rats, Wistar
20.
J Neurosci ; 34(13): 4445-52, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24671991

ABSTRACT

The rodent transient receptor potential ankyrin-1 (TRPA1) channel has been hypothesized to serve as a temperature sensor for thermoregulation in the cold. We tested this hypothesis by using deletion of the Trpa1 gene in mice and pharmacological blockade of the TRPA1 channel in rats. In both Trpa1(-/-) and Trpa1(+/+) mice, severe cold exposure (8°C) resulted in decreases of skin and deep body temperatures to ∼8°C and 13°C, respectively, both temperatures being below the reported 17°C threshold temperature for TRPA1 activation. Under these conditions, Trpa1(-/-) mice had the same dynamics of body temperature as Trpa1(+/+) mice and showed no weakness in the tail skin vasoconstriction response or thermogenic response to cold. In rats, the effects of pharmacological blockade were studied by using two chemically unrelated TRPA1 antagonists: the highly potent and selective compound A967079, which had been characterized earlier, and the relatively new compound 43 ((4R)-1,2,3,4-tetrahydro-4-[3-(3-methoxypropoxy)phenyl]-2-thioxo-5H-indeno[1,2-d]pyrimidin-5-one), which we further characterized in the present study and found to be highly potent (IC50 against cold of ∼8 nm) and selective. Intragastric administration of either antagonist at 30 mg/kg before severe (3°C) cold exposure did not affect the thermoregulatory responses (deep body and tail skin temperatures) of rats, even though plasma concentrations of both antagonists well exceeded their IC50 value at the end of the experiment. In the same experimental setup, blocking the melastatin-8 (TRPM8) channel with AMG2850 (30 mg/kg) attenuated cold-defense mechanisms and led to hypothermia. We conclude that TRPA1 channels do not drive autonomic thermoregulatory responses to cold in rodents.


Subject(s)
Autonomic Nervous System/physiology , Body Temperature Regulation/genetics , Intracellular Signaling Peptides and Proteins/physiology , Thermosensing/genetics , Animals , Body Temperature Regulation/drug effects , CHO Cells , Cold Temperature , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HSP90 Heat-Shock Proteins , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Transgenic , Oximes/blood , Oximes/pharmacology , Pain/chemically induced , Pain/drug therapy , Rats , Rats, Sprague-Dawley , Rats, Wistar , Skin Temperature/drug effects , TRPM Cation Channels/antagonists & inhibitors , Thermosensing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...