Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Am J Physiol Endocrinol Metab ; 322(1): E10-E23, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34779255

RESUMO

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.


Assuntos
Temperatura Corporal/efeitos dos fármacos , Colecistocinina/administração & dosagem , Ciclo-Oxigenase 2/metabolismo , Hipertermia/induzido quimicamente , Hipertermia/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Anorexia/induzido quimicamente , Benzodiazepinas/administração & dosagem , Regulação da Temperatura Corporal/efeitos dos fármacos , Colecistocinina/efeitos adversos , Inibidores de Ciclo-Oxigenase 2/administração & dosagem , Modelos Animais de Doenças , Ingestão de Alimentos/efeitos dos fármacos , Febre/induzido quimicamente , Febre/tratamento farmacológico , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Injeções Intraventriculares , Lipopolissacarídeos/efeitos adversos , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Receptor de Colecistocinina B/antagonistas & inibidores , Resultado do Tratamento
2.
J Neurosci ; 37(29): 6956-6971, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28630253

RESUMO

In the past, we showed that large electrolytic lesions of the dorsomedial hypothalamus (DMH) promoted hypothermia in cold-exposed restrained rats, but attenuated hypothermia in rats challenged with a high dose of bacterial lipopolysaccharide (LPS) in a thermogradient apparatus. The goal of this study was to identify the thermoeffector mechanisms and DMH representation of the two phenomena and thus to understand how the same lesion could produce two opposite effects on body temperature. We found that the permissive effect of large electrolytic DMH lesions on cold-induced hypothermia was due to suppressed thermogenesis. DMH-lesioned rats also could not develop fever autonomically: they did not increase thermogenesis in response to a low, pyrogenic dose of LPS (10 µg/kg, i.v.). In contrast, changes in thermogenesis were uninvolved in the attenuation of the hypothermic response to a high, shock-inducing dose of LPS (5000 µg/kg, i.v.); this attenuation was due to a blockade of cold-seeking behavior. To compile DMH maps for the autonomic cold defense and for the cold-seeking response to LPS, we studied rats with small thermal lesions in different parts of the DMH. Cold thermogenesis had the highest representation in the dorsal hypothalamic area. Cold seeking was represented by a site at the ventral border of the dorsomedial nucleus. Because LPS causes both fever and hypothermia, we originally thought that the DMH contained a single thermoregulatory site that worked as a fever-hypothermia switch. Instead, we have found two separate sites: one that drives thermogenesis and the other, previously unknown, that drives inflammation-associated cold seeking.SIGNIFICANCE STATEMENT Cold-seeking behavior is a life-saving response that occurs in severe systemic inflammation. We studied this behavior in rats with lesions in the dorsomedial hypothalamus (DMH) challenged with a shock-inducing dose of bacterial endotoxin. We built functional maps of the DMH and found the strongest representation of cold-seeking behavior at the ventral border of the dorsomedial nucleus. We also built maps for cold-induced thermogenesis in unanesthetized rats and found the dorsal hypothalamic area to be its main representation site. Our work identifies the neural substrate of cold-seeking behavior in systemic inflammation and expands the functional topography of the DMH, a structure that modulates autonomic, endocrine, and behavioral responses and is a potential therapeutic target in anxiety and panic disorders.


Assuntos
Comportamento Exploratório , Hipotálamo/fisiopatologia , Hipotermia/etiologia , Hipotermia/fisiopatologia , Inflamação/fisiopatologia , Termogênese , Animais , Comportamento Animal , Temperatura Baixa/efeitos adversos , Estado de Consciência , Imageamento Tridimensional/métodos , Imageamento por Ressonância Magnética/métodos , Masculino , Rede Nervosa/fisiopatologia , Vias Neurais/fisiopatologia , Ratos , Ratos Wistar
3.
Anesthesiology ; 127(5): 813-823, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28806222

RESUMO

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.


Assuntos
Analgésicos Opioides/uso terapêutico , Anestesia/efeitos adversos , Hipotermia/induzido quimicamente , Hipotermia/prevenção & controle , Dor Pós-Operatória/tratamento farmacológico , Canais de Cátion TRPV/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Benzotiazóis/farmacologia , Benzotiazóis/uso terapêutico , Relação Dose-Resposta a Droga , Feminino , Hipotermia/patologia , Indazóis/farmacologia , Indazóis/uso terapêutico , Masculino , Camundongos , Camundongos Knockout , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Dor Pós-Operatória/patologia , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Ureia/análogos & derivados , Ureia/farmacologia , Ureia/uso terapêutico
4.
J Neurosci ; 34(13): 4445-52, 2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24671991

RESUMO

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.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Regulação da Temperatura Corporal/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Sensação Térmica/genética , Animais , Regulação da Temperatura Corporal/efeitos dos fármacos , Células CHO , Temperatura Baixa , Cricetulus , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP90 , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Transgênicos , Oximas/sangue , Oximas/farmacologia , Dor/induzido quimicamente , Dor/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Temperatura Cutânea/efeitos dos fármacos , Canais de Cátion TRPM/antagonistas & inibidores , Sensação Térmica/efeitos dos fármacos
5.
Front Neuroendocrinol ; 34(3): 211-27, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23872331

RESUMO

Despite affecting millions of individuals, the etiology of hot flushes remains unknown. Here we review the physiology of hot flushes, CNS pathways regulating heat-dissipation effectors, and effects of estrogen on thermoregulation in animal models. Based on the marked changes in hypothalamic kisspeptin, neurokinin B and dynorphin (KNDy) neurons in postmenopausal women, we hypothesize that KNDy neurons play a role in the mechanism of flushes. In the rat, KNDy neurons project to preoptic thermoregulatory areas that express the neurokinin 3 receptor (NK3R), the primary receptor for NKB. Furthermore, activation of NK3R in the median preoptic nucleus, part of the heat-defense pathway, reduces body temperature. Finally, ablation of KNDy neurons reduces cutaneous vasodilatation and partially blocks the effects of estrogen on thermoregulation. These data suggest that arcuate KNDy neurons relay estrogen signals to preoptic structures regulating heat-dissipation effectors, supporting the hypothesis that KNDy neurons participate in the generation of flushes.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Dinorfinas/fisiologia , Fogachos/fisiopatologia , Hipotálamo/metabolismo , Kisspeptinas/fisiologia , Neurocinina B/fisiologia , Animais , Regulação da Temperatura Corporal/efeitos dos fármacos , Estradiol/farmacologia , Ciclo Estral/efeitos dos fármacos , Feminino , Humanos , Hormônio Luteinizante/metabolismo , Modelos Biológicos , Neurônios/fisiologia , Ovariectomia , Pós-Menopausa/fisiologia , Área Pré-Óptica/metabolismo , Ratos , Receptores da Neurocinina-3/metabolismo , Transdução de Sinais , Pele/irrigação sanguínea , Cauda/irrigação sanguínea , Vasodilatação
6.
J Neurosci ; 32(6): 2086-99, 2012 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-22323721

RESUMO

We studied N-(2-aminoethyl)-N-(4-(benzyloxy)-3-methoxybenzyl)thiophene-2-carboxamide hydrochloride (M8-B), a selective and potent antagonist of the transient receptor potential melastatin-8 (TRPM8) channel. In vitro, M8-B blocked cold-induced and TRPM8-agonist-induced activation of rat, human, and murine TRPM8 channels, including those on primary sensory neurons. In vivo, M8-B decreased deep body temperature (T(b)) in Trpm8(+/+) mice and rats, but not in Trpm8(-/-) mice, thus suggesting an on-target action. Intravenous administration of M8-B was more effective in decreasing T(b) in rats than intrathecal or intracerebroventricular administration, indicating a peripheral action. M8-B attenuated cold-induced c-Fos expression in the lateral parabrachial nucleus, thus indicating a site of action within the cutaneous cooling neural pathway to thermoeffectors, presumably on sensory neurons. A low intravenous dose of M8-B did not affect T(b) at either a constantly high or a constantly low ambient temperature (T(a)), but the same dose readily decreased T(b) if rats were kept at a high T(a) during the M8-B infusion and transferred to a low T(a) immediately thereafter. These data suggest that both a successful delivery of M8-B to the skin (high cutaneous perfusion) and the activation of cutaneous TRPM8 channels (by cold) are required for the hypothermic action of M8-B. At tail-skin temperatures <23°C, the magnitude of the M8-B-induced decrease in T(b) was inversely related to skin temperature, thus suggesting that M8-B blocks thermal (cold) activation of TRPM8. M8-B affected all thermoeffectors studied (thermopreferendum, tail-skin vasoconstriction, and brown fat thermogenesis), thus suggesting that TRPM8 is a universal cold receptor in the thermoregulation system.


Assuntos
Temperatura Corporal/fisiologia , Temperatura Baixa , Gânglios Espinais/fisiologia , Estremecimento/fisiologia , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/deficiência , Animais , Sistema Nervoso Autônomo/efeitos dos fármacos , Sistema Nervoso Autônomo/fisiologia , Comportamento Animal/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Gânglios Espinais/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Preparações Farmacêuticas/administração & dosagem , Ratos , Ratos Wistar , Estremecimento/efeitos dos fármacos , Tiofenos/farmacologia
8.
Temperature (Austin) ; 10(1): 121-135, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37187830

RESUMO

Thermoregulatory behaviors are powerful effectors for core body temperature (Tc) regulation. We evaluated the involvement of afferent fibers ascending through the dorsal portion of the lateral funiculus (DLF) of the spinal cord in "spontaneous" thermal preference and thermoregulatory behaviors induced by thermal and pharmacological stimuli in a thermogradient apparatus. In adult Wistar rats, the DLF was surgically severed at the first cervical vertebra bilaterally. The functional effectiveness of funiculotomy was verified by the increased latency of tail-flick responses to noxious cold (-18°C) and heat (50°C). In the thermogradient apparatus, funiculotomized rats showed a higher variability of their preferred ambient temperature (Tpr) and, consequently, increased Tc fluctuations, as compared to sham-operated rats. The cold-avoidance (warmth-seeking) response to moderate cold (whole-body exposure to ~17°C) or epidermal menthol (an agonist of the cold-sensitive TRPM8 channel) was attenuated in funiculotomized rats, as compared to sham-operated rats, and so was the Tc (hyperthermic) response to menthol. In contrast, the warmth-avoidance (cold-seeking) and Tc responses of funiculotomized rats to mild heat (exposure to ~28°C) or intravenous RN-1747 (an agonist of the warmth-sensitive TRPV4; 100 µg/kg) were unaffected. We conclude that DLF-mediated signals contribute to driving spontaneous thermal preference, and that attenuation of these signals is associated with decreased precision of Tc regulation. We further conclude that thermally and pharmacologically induced changes in thermal preference rely on neural, presumably afferent, signals that travel in the spinal cord within the DLF. Signals conveyed by the DLF are important for cold-avoidance behaviors but make little contribution to heat-avoidance responses.

9.
Temperature (Austin) ; 10(1): 136-154, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37187834

RESUMO

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.

10.
J Neurosci ; 31(5): 1721-33, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21289181

RESUMO

This study aimed at determining the thermoregulatory phenotype of mice lacking transient receptor potential vanilloid-1 (TRPV1) channels. We used Trpv1 knockout (KO) mice and their genetically unaltered littermates to study diurnal variations in deep body temperature (T(b)) and thermoeffector activities under basal conditions, as well as thermoregulatory responses to severe heat and cold. Only subtle alterations were found in the basal T(b) of Trpv1 KO mice or in their T(b) responses to thermal challenges. The main thermoregulatory abnormality of Trpv1 KO mice was a different pattern of thermoeffectors used to regulate T(b). On the autonomic side, Trpv1 KO mice were hypometabolic (had a lower oxygen consumption) and hypervasoconstricted (had a lower tail skin temperature). In agreement with the enhanced skin vasoconstriction, Trpv1 KO mice had a higher thermoneutral zone. On the behavioral side, Trpv1 KO mice preferred a lower ambient temperature and expressed a higher locomotor activity. Experiments with pharmacological TRPV1 agonists (resiniferatoxin and anandamide) and a TRPV1 antagonist (AMG0347) confirmed that TRPV1 channels located outside the brain tonically inhibit locomotor activity. With age (observed for up to 14 months), the body mass of Trpv1 KO mice exceeded that of controls, sometimes approaching 60 g. In summary, Trpv1 KO mice possess a distinct thermoregulatory phenotype, which is coupled with a predisposition to age-associated overweight and includes hypometabolism, enhanced skin vasoconstriction, decreased thermopreferendum, and hyperkinesis. The latter may be one of the primary deficiencies in Trpv1 KO mice. We propose that TRPV1-mediated signals from the periphery tonically suppress the general locomotor activity.


Assuntos
Envelhecimento/metabolismo , Regulação da Temperatura Corporal/genética , Hipercinese/metabolismo , Sobrepeso/metabolismo , Canais de Cátion TRPV/deficiência , Acrilamidas/farmacologia , Animais , Ácidos Araquidônicos/farmacologia , Temperatura Corporal/genética , Temperatura Baixa , Diterpenos/farmacologia , Endocanabinoides , Feminino , Temperatura Alta , Hipercinese/genética , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Testes Neuropsicológicos , Consumo de Oxigênio , Fenótipo , Reação em Cadeia da Polimerase , Alcamidas Poli-Insaturadas/farmacologia , Piridinas/farmacologia , Pele/irrigação sanguínea , Temperatura Cutânea/genética , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/genética , Vasoconstrição
11.
Am J Physiol Regul Integr Comp Physiol ; 302(12): R1372-83, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22513748

RESUMO

The natural switch from fever to hypothermia observed in the most severe cases of systemic inflammation is a phenomenon that continues to puzzle clinicians and scientists. The present study was the first to evaluate in direct experiments how the development of hypothermia vs. fever during severe forms of systemic inflammation impacts the pathophysiology of this malady and mortality rates in rats. Following administration of bacterial lipopolysaccharide (LPS; 5 or 18 mg/kg) or of a clinical Escherichia coli isolate (5 × 10(9) or 1 × 10(10) CFU/kg), hypothermia developed in rats exposed to a mildly cool environment, but not in rats exposed to a warm environment; only fever was revealed in the warm environment. Development of hypothermia instead of fever suppressed endotoxemia in E. coli-infected rats, but not in LPS-injected rats. The infiltration of the lungs by neutrophils was similarly suppressed in E. coli-infected rats of the hypothermic group. These potentially beneficial effects came with costs, as hypothermia increased bacterial burden in the liver. Furthermore, the hypotensive responses to LPS or E. coli were exaggerated in rats of the hypothermic group. This exaggeration, however, occurred independently of changes in inflammatory cytokines and prostaglandins. Despite possible costs, development of hypothermia lessened abdominal organ dysfunction and reduced overall mortality rates in both the E. coli and LPS models. By demonstrating that naturally occurring hypothermia is more advantageous than fever in severe forms of aseptic (LPS-induced) or septic (E. coli-induced) systemic inflammation, this study provides new grounds for the management of this deadly condition.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Escherichia coli , Febre/fisiopatologia , Hipotermia/fisiopatologia , Inflamação/fisiopatologia , Lipopolissacarídeos , Animais , Temperatura Corporal/fisiologia , Febre/induzido quimicamente , Hipotermia/induzido quimicamente , Inflamação/induzido quimicamente , Masculino , Ratos , Ratos Wistar
12.
Pharmacol Rev ; 61(3): 228-61, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19749171

RESUMO

The development of antagonists of the transient receptor potential vanilloid-1 (TRPV1) channel as pain therapeutics has revealed that these compounds cause hyperthermia in humans. This undesirable on-target side effect has triggered a surge of interest in the role of TRPV1 in thermoregulation and revived the hypothesis that TRPV1 channels serve as thermosensors. We review literature data on the distribution of TRPV1 channels in the body and on thermoregulatory responses to TRPV1 agonists and antagonists. We propose that two principal populations of TRPV1-expressing cells have connections with efferent thermoeffector pathways: 1) first-order sensory (polymodal), glutamatergic dorsal-root (and possibly nodose) ganglia neurons that innervate the abdominal viscera and 2) higher-order sensory, glutamatergic neurons presumably located in the median preoptic hypothalamic nucleus. We further hypothesize that all thermoregulatory responses to TRPV1 agonists and antagonists and thermoregulatory manifestations of TRPV1 desensitization stem from primary actions on these two neuronal populations. Agonists act primarily centrally on population 2; antagonists act primarily peripherally on population 1. We analyze what roles TRPV1 might play in thermoregulation and conclude that this channel does not serve as a thermosensor, at least not under physiological conditions. In the hypothalamus, TRPV1 channels are inactive at common brain temperatures. In the abdomen, TRPV1 channels are tonically activated, but not by temperature. However, tonic activation of visceral TRPV1 by nonthermal factors suppresses autonomic cold-defense effectors and, consequently, body temperature. Blockade of this activation by TRPV1 antagonists disinhibits thermoeffectors and causes hyperthermia. Strategies for creating hyperthermia-free TRPV1 antagonists are outlined. The potential physiological and pathological significance of TRPV1-mediated thermoregulatory effects is discussed.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Canais de Cátion TRPV/fisiologia , Animais , Humanos , Modelos Biológicos , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/antagonistas & inibidores
13.
J Neurosci ; 30(4): 1435-40, 2010 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-20107070

RESUMO

Transient receptor potential vanilloid-1 (TRPV1) antagonists are widely viewed as next-generation pain therapeutics. However, these compounds cause hyperthermia, a serious side effect. TRPV1 antagonists differentially block three modes of TRPV1 activation: by heat, protons, and chemical ligands (e.g., capsaicin). We asked what combination of potencies in these three modes of TRPV1 activation corresponds to the lowest potency of a TRPV1 antagonist to cause hyperthermia. We studied hyperthermic responses of rats, mice, and guinea pigs to eight TRPV1 antagonists with different pharmacological profiles and used mathematical modeling to find a relative contribution of the blockade of each activation mode to the development of hyperthermia. We found that the hyperthermic effect has the highest sensitivity to the extent of TRPV1 blockade in the proton mode (0.43 to 0.65) with no to moderate sensitivity in the capsaicin mode (-0.01 to 0.34) and no sensitivity in the heat mode (0.00 to 0.01). We conclude that hyperthermia-free TRPV1 antagonists do not block TRPV1 activation by protons, even if they are potent blockers of the heat mode, and that decreasing the potency to block the capsaicin mode may further decrease the potency to cause hyperthermia.


Assuntos
Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Febre/induzido quimicamente , Febre/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Animais , Capsaicina/antagonistas & inibidores , Sistema Nervoso Central/fisiopatologia , Simulação por Computador , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Febre/fisiopatologia , Cobaias , Temperatura Alta/efeitos adversos , Masculino , Camundongos , Camundongos Knockout , Neurofarmacologia/métodos , Terapia com Prótons , Ratos , Ratos Wistar , Fármacos do Sistema Sensorial/farmacologia
14.
J Physiol ; 589(Pt 9): 2415-31, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21486787

RESUMO

Hypothermia occurs in the most severe cases of systemic inflammation, but the mechanisms involved are poorly understood. This study evaluated whether the hypothermic response to bacterial lipopolysaccharide (LPS) is modulated by the endocannabinoid anandamide(AEA) and its receptors: cannabinoid-1 (CB1), cannabinoid-2 (CB2) and transient receptor potential vanilloid-1 (TRPV1). In rats exposed to an ambient temperature of 22◦C, a moderate dose of LPS (25 - 100 µg kg−1 I.V.) induced a fall in body temperature with a nadir at ∼100 minpostinjection. This response was not affected by desensitization of intra-abdominal TRPV1 receptors with resiniferatoxin (20 µg kg - 1 I.P.), by systemic TRPV1 antagonism with capsazepine(40mg kg−1 I.P.), or by systemic CB2 receptor antagonism with SR144528 (1.4 mg kg−1 I.P.).However, CB1 receptor antagonism by rimonabant (4.6mg kg−1 I.P.) or SLV319 (15mg kg−1 I.P.)blocked LPS hypothermia. The effect of rimonabant was further studied. Rimonabant blocked LPS hypothermia when administered I.C.V. at a dose (4.6 µg) that was too low to produce systemic effects. The blockade of LPS hypothermia by I.C.V. rimonabant was associated with suppression of the circulating level of tumour necrosis factor-α. In contrast to rimonabant,the I.C.V. administration of AEA (50 µg) enhanced LPS hypothermia. Importantly, I.C.V. AEAdid not evoke hypothermia in rats not treated with LPS, thus indicating that AEA modulates LPS-activated pathways in the brain rather than thermo effector pathways. In conclusion, the present study reveals a novel, critical role of brain CB1 receptors in LPS hypothermia. Brain CB1 receptors may constitute a new therapeutic target in systemic inflammation and sepsis.


Assuntos
Regulação da Temperatura Corporal , Encéfalo/metabolismo , Hipotermia/metabolismo , Lipopolissacarídeos , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Canais de Cátion TRPV/metabolismo , Análise de Variância , Animais , Ácidos Araquidônicos/metabolismo , Regulação da Temperatura Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Canfanos/administração & dosagem , Capsaicina/administração & dosagem , Capsaicina/análogos & derivados , Modelos Animais de Doenças , Diterpenos/administração & dosagem , Endocanabinoides , Feminino , Hipotermia/induzido quimicamente , Hipotermia/fisiopatologia , Hipotermia/prevenção & controle , Injeções Intraperitoneais , Injeções Intravenosas , Injeções Intraventriculares , Masculino , Piperidinas/administração & dosagem , Alcamidas Poli-Insaturadas/metabolismo , Pirazóis/administração & dosagem , Ratos , Ratos Long-Evans , Ratos Wistar , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/antagonistas & inibidores , Rimonabanto , Transdução de Sinais , Sulfonamidas/administração & dosagem , Canais de Cátion TRPV/antagonistas & inibidores , Fatores de Tempo
15.
J Neurotrauma ; 38(19): 2772-2776, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34002636

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas/mortalidade , Lesões Encefálicas Traumáticas/terapia , Hipotermia Induzida , Humanos , Avaliação de Resultados em Cuidados de Saúde , Seleção de Pacientes
16.
J Trauma ; 69(2): 308-12, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20699738

RESUMO

BACKGROUND: There is a high percentage of smokers among trauma patients. Cigarette smoking has been associated with the development of acute lung injury and the adult respiratory distress syndrome in critically ill patients. It is also known that nicotine exerts immunosuppressive and anti-inflammatory effects with chronic use. Trauma patients who are smokers usually go through acute nicotine withdrawal after the traumatic event and during their stay in ICU. How the smoking status and acute nicotine withdrawal affect outcomes after trauma is unknown. This question was addressed in this study by analyzing the incidence of sepsis, septic shock and multiple organ dysfunction syndrome, and other outcomes in smoking and nonsmoking trauma patients. METHODS: A retrospective cohort of trauma patients who met the criteria was randomly selected from the trauma registry. Individual charts were reviewed to confirm documented smoking status. Criteria for selection included the following: Injury Severity Score >or=20, age 18 to 65 years, hospital length of stay >72 hours. Patients with COPD/emphysema, diabetes mellitus, cardiac disease, malignancy, pregnancy, or steroid use were excluded. RESULTS: Overall, 327 patient charts were reviewed: 156 smokers and 171 nonsmokers. Men outnumbered women in the smoking group fourfold (p = 0.003 versus nonsmokers). Age, Injury Severity Score, the presence of shock on admission, the type of trauma (blunt or penetrating), ICU and hospital length of stay, and the duration of ventilator support were similar between smokers and nonsmokers. There were no differences in the incidence of sepsis, pneumonia, adult respiratory distress syndrome, or multiple organ dysfunction syndrome. Mortality was low (1.2% in smokers; 0.6% in nonsmokers) and did not differ significantly between the groups. CONCLUSIONS: The smoking status plays a minimal role in the outcome of healthy trauma patients. This suggests that the acute nicotine withdrawal that usually occurs in critically ill patients has no clinically significant implications after injury.


Assuntos
Causas de Morte , Insuficiência de Múltiplos Órgãos/epidemiologia , Insuficiência Respiratória/epidemiologia , Sepse/epidemiologia , Fumar/epidemiologia , Ferimentos e Lesões/mortalidade , Adolescente , Adulto , Idoso , Arizona/epidemiologia , Causalidade , Estudos de Coortes , Comorbidade , Cuidados Críticos , Feminino , Mortalidade Hospitalar/tendências , Humanos , Escala de Gravidade do Ferimento , Unidades de Terapia Intensiva , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/etiologia , Probabilidade , Prognóstico , Valores de Referência , Sistema de Registros , Insuficiência Respiratória/etiologia , Estudos Retrospectivos , Sepse/etiologia , Fumar/efeitos adversos , Análise de Sobrevida , Ferimentos e Lesões/diagnóstico , Ferimentos e Lesões/terapia , Adulto Jovem
17.
Pharmacol Ther ; 208: 107474, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31926897

RESUMO

Antagonists of the transient receptor potential vanilloid-1 (TRPV1) channel alter body temperature (Tb) in laboratory animals and humans: most cause hyperthermia; some produce hypothermia; and yet others have no effect. TRPV1 can be activated by capsaicin (CAP), protons (low pH), and heat. First-generation (polymodal) TRPV1 antagonists potently block all three TRPV1 activation modes. Second-generation (mode-selective) TRPV1 antagonists potently block channel activation by CAP, but exert different effects (e.g., potentiation, no effect, or low-potency inhibition) in the proton mode, heat mode, or both. Based on our earlier studies in rats, only one mode of TRPV1 activation - by protons - is involved in thermoregulatory responses to TRPV1 antagonists. In rats, compounds that potently block, potentiate, or have no effect on proton activation cause hyperthermia, hypothermia, or no effect on Tb, respectively. A Tb response occurs when a TRPV1 antagonist blocks (in case of hyperthermia) or potentiates (hypothermia) the tonic TRPV1 activation by protons somewhere in the trunk, perhaps in muscles, and - via the acido-antithermogenic and acido-antivasoconstrictor reflexes - modulates thermogenesis and skin vasoconstriction. In this work, we used a mathematical model to analyze Tb data from human clinical trials of TRPV1 antagonists. The analysis suggests that, in humans, the hyperthermic effect depends on the antagonist's potency to block TRPV1 activation not only by protons, but also by heat, while the CAP activation mode is uninvolved. Whereas in rats TRPV1 drives thermoeffectors by mediating pH signals from the trunk, but not Tb signals, our analysis suggests that TRPV1 mediates both pH and thermal signals driving thermoregulation in humans. Hence, in humans (but not in rats), TRPV1 is likely to serve as a thermosensor of the thermoregulation system. We also conducted a meta-analysis of Tb data from human trials and found that polymodal TRPV1 antagonists (ABT-102, AZD1386, and V116517) increase Tb, whereas the mode-selective blocker NEO6860 does not. Several strategies of harnessing the thermoregulatory effects of TRPV1 antagonists in humans are discussed.


Assuntos
Hipertermia/induzido quimicamente , Modelos Biológicos , Canais de Cátion TRPV/antagonistas & inibidores , Animais , Desenvolvimento de Medicamentos , Humanos
18.
Am J Physiol Regul Integr Comp Physiol ; 297(2): R485-94, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19515980

RESUMO

Systemic inflammation is associated with either fever or hypothermia. Fever, a response to mild systemic inflammation, is mediated by cyclooxygenase (COX)-2 and not by COX-1. However, it is still disputed whether COX-2, COX-1, neither, or both mediate(s) responses to severe systemic inflammation, and, in particular, the hypothermic response. We compared the effects of SC-236 (COX-2 inhibitor) and SC-560 (COX-1 inhibitor) on the deep body temperature (T(b)) of rats injected with a lower (10 microg/kg i.v.) or higher (1,000 microg/kg i.v.) dose of LPS at different ambient temperatures (T(a)s). At a neutral T(a) (30 degrees C), the rats responded to LPS with a polyphasic fever (lower dose) or a brief hypothermia followed by fever (higher dose). SC-236 (2.5 mg/kg i.v.) blocked the fever induced by either LPS dose, whereas SC-560 (5 mg/kg i.v.) altered neither the febrile response to the lower LPS dose nor the fever component of the response to the higher dose. However, SC-560 blocked the initial hypothermia caused by the higher LPS dose. At a subneutral T(a) (22 degrees C), the rats responded to LPS with early (70-90 min, nadir) dose-dependent hypothermia. The hypothermic response to either dose was enhanced by SC-236 but blocked by SC-560. The hypothermic response to the higher LPS dose was associated with a fall in arterial blood pressure. This hypotensive response was attenuated by either SC-236 or SC-560. At the onset of LPS-induced hypothermia and hypotension, the functional activity of the COX-1 pathway (COX-1-mediated PGE(2) synthesis ex vivo) increased in the spleen but not liver, lung, kidney, or brain. The expression of splenic COX-1 was unaffected by LPS. We conclude that COX-1, but not COX-2, mediates LPS hypothermia, and that both COX isoforms are required for LPS hypotension.


Assuntos
Ciclo-Oxigenase 1/fisiologia , Ciclo-Oxigenase 2/fisiologia , Hipotermia/induzido quimicamente , Hipotermia/metabolismo , Lipopolissacarídeos/farmacologia , Estruturas Animais/efeitos dos fármacos , Estruturas Animais/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Regulação da Temperatura Corporal/efeitos dos fármacos , Regulação da Temperatura Corporal/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Expressão Gênica/efeitos dos fármacos , Hipotermia/fisiopatologia , Masculino , Pirazóis/farmacologia , Ratos , Ratos Wistar , Baço/efeitos dos fármacos , Baço/metabolismo , Sulfonamidas/farmacologia , Temperatura
19.
PLoS Biol ; 4(9): e284, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16933973

RESUMO

All phases of lipopolysaccharide (LPS)-induced fever are mediated by prostaglandin (PG) E2. It is known that the second febrile phase (which starts at approximately 1.5 h post-LPS) and subsequent phases are mediated by PGE2 that originated in endotheliocytes and perivascular cells of the brain. However, the location and phenotypes of the cells that produce PGE2 triggering the first febrile phase (which starts at approximately 0.5 h) remain unknown. By studying PGE2 synthesis at the enzymatic level, we found that it was activated in the lung and liver, but not in the brain, at the onset of the first phase of LPS fever in rats. This activation involved phosphorylation of cytosolic phospholipase A2 (cPLA2) and transcriptional up-regulation of cyclooxygenase (COX)-2. The number of cells displaying COX-2 immunoreactivity surged in the lung and liver (but not in the brain) at the onset of fever, and the majority of these cells were identified as macrophages. When PGE2 synthesis in the periphery was activated, the concentration of PGE2 increased both in the venous blood (which collects PGE2 from tissues) and arterial blood (which delivers PGE2 to the brain). Most importantly, neutralization of circulating PGE2 with an anti-PGE2 antibody both delayed and attenuated LPS fever. It is concluded that fever is initiated by circulating PGE2 synthesized by macrophages of the LPS-processing organs (lung and liver) via phosphorylation of cPLA2 and transcriptional up-regulation of COX-2. Whether PGE2 produced at the level of the blood-brain barrier also contributes to the development of the first phase remains to be clarified.


Assuntos
Febre/induzido quimicamente , Febre/metabolismo , Lipopolissacarídeos/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Febre/fisiopatologia , Regulação Enzimológica da Expressão Gênica , Fígado/citologia , Fígado/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Macrófagos/metabolismo , Masculino , Ratos , Ratos Long-Evans , Transdução de Sinais , Regulação para Cima
20.
Prog Lipid Res ; 46(2): 89-107, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17275915

RESUMO

In addition to playing a central role in energy homeostasis, leptin is also an important player in the inflammatory response. Systemic inflammation is accompanied by fever (less severe cases) or hypothermia (more severe cases). In leptin-irresponsive mutants, the hypothermia of systemic inflammation is exaggerated, presumably due to the enhanced production and cryogenic action of tumor necrosis factor (TNF)-alpha. Mechanisms that exaggerate hypothermia can also attenuate fever, particularly in a cool environment. Another common manifestation of systemic inflammation is behavioral depression. Along with the production of interleukin (IL)-1beta, this manifestation is exaggerated in leptin-irresponsive mutants. The enhanced production of TNF-alpha and IL-1beta may be due, at least in part, to insufficient activation of the anti-inflammatory hypothalamo-pituitary-adrenal axis by immune stimuli in the absence of leptin signaling. In experimental animals and humans that are responsive to leptin, suppression of leptin production under conditions of negative energy balance (e.g., fasting) can exaggerate both hypothermia and behavioral depression. Since these manifestations aid energy conservation, exaggeration of these manifestations under conditions of negative energy balance is likely to be beneficial.


Assuntos
Inflamação/metabolismo , Leptina/metabolismo , Animais , Homeostase , Humanos , Modelos Genéticos , Receptores para Leptina/química , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA