Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Mol Metab ; 84: 101946, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38657735

RESUMO

Our circadian world shapes much of metabolic physiology. In mice ∼40% of the light and ∼80% of the dark phase time is characterized by bouts of increased energy expenditure (EE). These ultradian bouts have a higher body temperature (Tb) and thermal conductance and contain virtually all of the physical activity and awake time. Bout status is a better classifier of mouse physiology than photoperiod, with ultradian bouts superimposed on top of the circadian light/dark cycle. We suggest that the primary driver of ultradian bouts is a brain-initiated transition to a higher defended Tb of the active/awake state. Increased energy expenditure from brown adipose tissue, physical activity, and cardiac work combine to raise Tb from the lower defended Tb of the resting/sleeping state. Thus, unlike humans, much of mouse metabolic physiology is episodic with large ultradian increases in EE and Tb that correlate with the active/awake state and are poorly aligned with circadian cycling.


Assuntos
Temperatura Corporal , Ritmo Circadiano , Metabolismo Energético , Fotoperíodo , Ritmo Ultradiano , Animais , Camundongos , Ritmo Ultradiano/fisiologia , Metabolismo Energético/fisiologia , Ritmo Circadiano/fisiologia , Temperatura Corporal/fisiologia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/fisiologia , Vigília/fisiologia , Sono/fisiologia
2.
PLoS One ; 18(10): e0292610, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37812612

RESUMO

OBJECTIVE: Otopetrin 1 (OTOP1) is a proton channel that is highly expressed in brown adipose tissue. We examined the physiology of Otop1-/- mice, which lack functional OTOP1. METHODS: Mice were studied by indirect calorimetry and telemetric ambulatory body temperature monitoring. Mitochondrial function was measured as oxygen consumption and extracellular acidification. RESULTS: Otop1-/- mice had similar body temperatures as control mice at baseline and in response to cold and hot ambient temperatures. However, in response to fasting the Otop1-/- mice exhibited an exaggerated hypothermia and hypometabolism. Similarly, in ex vivo tests of Otop1-/- brown adipose tissue mitochondrial function, there was no change in baseline oxygen consumption, but the oxygen consumption was reduced after maximal uncoupling with FCCP and increased upon stimulation with the ß3-adrenergic agonist CL316243. Mast cells also express Otop1, and Otop1-/- mice had intact, possibly greater hypothermia in response to mast cell activation by the adenosine A3 receptor agonist MRS5698. No increase in insulin resistance was observed in the Otop1-/- mice. CONCLUSIONS: Loss of OTOP1 does not change basal function of brown adipose tissue but affects stimulated responses.


Assuntos
Hipotermia , Animais , Camundongos , Tecido Adiposo Marrom , Temperatura Corporal , Regulação da Temperatura Corporal , Jejum , Camundongos Knockout
3.
Mol Metab ; 71: 101699, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36858190

RESUMO

OBJECTIVE: Physical activity is a major component of total energy expenditure (TEE) that exhibits extreme variability in mice. Our objective was to construct a general, physiology-based model of TEE to accurately quantify the energy cost of physical activity. METHODS: Spontaneous home cage physical activity, body temperature, TEE, and energy intake were measured with frequent sampling. The energy cost of activity was modeled considering six contributors to TEE (basal metabolic rate, thermic effect of food, body temperature, cold induced thermogenesis, physical activity, and body weight). An ambient temperature of 35 °C was required to remove the contribution from cold induced thermogenesis. Basal metabolic rate was adjusted for body temperature using a Q10 temperature coefficient. RESULTS: We developed a TEE model that robustly explains 70-80% of the variance in TEE at 35 °C while fitting only two parameters, the basal metabolic rate and the mass-specific energy cost per unit of physical activity, which averaged 60 cal/km/g body weight. In Ucp1-/- mice the activity cost was elevated by 60%, indicating inefficiency and increased muscle thermogenesis. The diurnal rhythm in TEE was quantitatively explained by the combined diurnal differences in physical activity, body temperature, and energy intake. Incorporating body temperature into human basal metabolic rate measurements significantly reduced the inter-individual variation. CONCLUSIONS: The physiology-based model of TEE allows quantifying the energy cost of physical activity. While applied here to mice, the model should be generally valid across species. Due to the effect of body temperature, we suggest that basal metabolic rate measurements be corrected to a reference body temperature, including in humans. Having an accurate cost of physical activity allows mechanistic dissection of disorders of energy homeostasis, including obesity.


Assuntos
Metabolismo Basal , Metabolismo Energético , Humanos , Animais , Camundongos , Metabolismo Energético/fisiologia , Peso Corporal/fisiologia , Metabolismo Basal/fisiologia , Obesidade , Termogênese/fisiologia
4.
Purinergic Signal ; 19(3): 551-564, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36781825

RESUMO

Some non-adenosinergic drugs are reported to also act through adenosine receptors (ARs). We used mouse hypothermia, which can be induced by agonism at any of the four ARs, as an in vivo screen for adenosinergic effects. An AR contribution was identified when a drug caused hypothermia in wild type mice that was diminished in mice lacking all four ARs (quadruple knockout, QKO). Alternatively, an adenosinergic effect was identified if a drug potentiated adenosine-induced hypothermia. Four drugs (dipyridamole, nimodipine, cilostazol, cyclosporin A) increased the hypothermia caused by adenosine. Dipyridamole and nimodipine probably achieved this by inhibition of adenosine clearance via ENT1. Two drugs (cannabidiol, canrenoate) did not cause hypothermia in wild type mice. Four other drugs (nifedipine, ranolazine, ketamine, ethanol) caused hypothermia, but the hypothermia was unchanged in QKO mice indicating non-adenosinergic mechanisms. Zinc chloride caused hypothermia and hypoactivity; the hypoactivity was blunted in the QKO mice. Interestingly, the antidepressant amitriptyline caused hypothermia in wild type mice that was amplified in the QKO mice. Thus, we have identified adenosine-related effects for some drugs, while other candidates do not affect adenosine signaling by this in vivo assay. The adenosine-modulating drugs could be considered for repurposing based on predicted effects on AR activation.


Assuntos
Adenosina , Hipotermia , Camundongos , Animais , Adenosina/farmacologia , Hipotermia/induzido quimicamente , Nimodipina/efeitos adversos , Receptores Purinérgicos P1 , Dipiridamol/efeitos adversos
5.
Mol Metab ; 53: 101332, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34478905

RESUMO

OBJECTIVE: To improve understanding of mouse energy homeostasis and its applicability to humans, we quantitated the effects of housing density on mouse thermal physiology in both sexes. METHODS: Littermate wild type and Brs3-null mice were single- or group- (three per cage) housed and studied by indirect calorimetry with continuous measurement of core body temperature, energy expenditure, physical activity, and food intake. RESULTS: At 23 °C, below thermoneutrality, single-housed males had a lower body temperature and unchanged metabolic rate compared to group-housed controls. In contrast, single-housed females maintained a similar body temperature to group-housed controls by increasing their metabolic rate. With decreasing ambient temperature below 27 °C, only group-housed mice decreased their heat conductance, likely due to huddling, thus interfering with the energy expenditure vs ambient temperature relationship described by Scholander. In a hot environment (35 °C), the single-housed mice were less heat stressed. Upon fasting, single-housed mice had larger reductions in body temperature, with male mice having more torpor episodes of similar duration and female mice having a similar number of torpor episodes that lasted longer. Qualitatively, the effects of housing density on thermal physiology of Brs3-null mice generally mimicked the effects in controls. CONCLUSIONS: Single housing is more sensitive than group housing for detecting thermal physiology phenotypes. Single housing increases heat loss and amplifies the effects of fasting or a cold environment. Male and female mice utilize different thermoregulatory strategies to respond to single housing.


Assuntos
Composição Corporal/fisiologia , Temperatura Corporal , Receptores da Bombesina/metabolismo , Temperatura , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores da Bombesina/deficiência
6.
eNeuro ; 8(4)2021.
Artigo em Inglês | MEDLINE | ID: mdl-34326065

RESUMO

Bombesin receptor subtype-3 (BRS3) is an orphan receptor that regulates energy homeostasis. We compared Brs3 driver mice with constitutive or inducible Cre recombinase activity. The constitutive BRS3-Cre mice show a reporter signal (Cre-dependent tdTomato) in the adult brain because of lineage tracing in the dentate gyrus, striatal patches, and indusium griseum, in addition to sites previously identified in the inducible BRS3-Cre mice (including hypothalamic and amygdala subregions, and parabrachial nucleus). We detected Brs3 reporter expression in the dentate gyrus at day 23 but not at postnatal day 1 or 5 months of age. Hypothalamic sites expressed reporter at all three time points, and striatal patches expressed Brs3 reporter at 1 day but not 5 months. Parabrachial nucleus Brs3 neurons project to the preoptic area, hypothalamus, amygdala, and thalamus. Both Cre recombinase insertions reduced Brs3 mRNA levels and BRS3 function, causing obesity phenotypes of different severity. These results demonstrate that driver mice should be characterized phenotypically and illustrate the need for knock-in strategies with less effect on the endogenous gene.


Assuntos
Integrases , Receptores da Bombesina , Animais , Encéfalo/metabolismo , Hipotálamo/metabolismo , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Receptores da Bombesina/metabolismo
7.
Cell Metab ; 33(7): 1389-1403.e6, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34038711

RESUMO

The preoptic area (POA) is a key brain region for regulation of body temperature (Tb), dictating thermogenic, cardiovascular, and behavioral responses that control Tb. Previously characterized POA neuronal populations all reduced Tb when activated. Using mice, we now identify POA neurons expressing bombesin-like receptor 3 (POABRS3) as a population whose activation increased Tb; inversely, acute inhibition of these neurons reduced Tb. POABRS3 neurons that project to either the paraventricular nucleus of the hypothalamus or the dorsomedial hypothalamus increased Tb, heart rate, and blood pressure via the sympathetic nervous system. Long-term inactivation of POABRS3 neurons caused increased Tb variability, overshooting both increases and decreases in Tb set point, with RNA expression profiles suggesting multiple types of POABRS3 neurons. Thus, POABRS3 neuronal populations regulate Tb and heart rate, contribute to cold defense, and fine-tune feedback control of Tb. These findings advance understanding of homeothermy, a defining feature of mammalian biology.


Assuntos
Regulação da Temperatura Corporal , Frequência Cardíaca , Neurônios/fisiologia , Área Pré-Óptica/metabolismo , Receptores da Bombesina/metabolismo , Animais , Temperatura Corporal/genética , Regulação da Temperatura Corporal/genética , Frequência Cardíaca/genética , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Área Pré-Óptica/citologia , Receptores da Bombesina/genética , Transdução de Sinais/genética , Sistema Nervoso Simpático/fisiologia , Termogênese/genética
8.
J Clin Invest ; 131(7)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33621215

RESUMO

The A3 adenosine receptor (A3AR) has emerged as a therapeutic target with A3AR agonists to tackle the global challenge of neuropathic pain, and investigation into its mode of action is essential for ongoing clinical development. Immune cell A3ARs, and their activation during pathology, modulate cytokine release. Thus, the use of immune cells as a cellular substrate for the pharmacological action of A3AR agonists is enticing, but unknown. The present study discovered that Rag-KO mice lacking T and B cells, as compared with WT mice, are insensitive to the anti-allodynic effects of A3AR agonists. Similar findings were observed in interleukin-10 and interleukin-10 receptor knockout mice. Adoptive transfer of CD4+ T cells from WT mice infiltrated the dorsal root ganglion (DRG) and restored A3AR agonist-mediated anti-allodynia in Rag-KO mice. CD4+ T cells from Adora3-KO or Il10-KO mice did not. Transfer of CD4+ T cells from WT mice, but not Il10-KO mice, into Il10-KO mice or Adora3-KO mice fully reinstated the anti-allodynic effects of A3AR activation. Notably, A3AR agonism reduced DRG neuron excitability when cocultured with CD4+ T cells in an IL-10-dependent manner. A3AR action on CD4+ T cells infiltrated in the DRG decreased phosphorylation of GluN2B-containing N-methyl-D-aspartate receptors at Tyr1472, a modification associated with regulating neuronal hypersensitivity. Our findings establish that activation of A3AR on CD4+ T cells to release IL-10 is required and sufficient evidence for the use of A3AR agonists as therapeutics.


Assuntos
Agonistas do Receptor A3 de Adenosina/farmacologia , Linfócitos T CD4-Positivos/imunologia , Gânglios Espinais/imunologia , Interleucina-10/imunologia , Neuralgia/tratamento farmacológico , Neurônios/imunologia , Receptor A3 de Adenosina/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Linfócitos T CD4-Positivos/patologia , Gânglios Espinais/patologia , Interleucina-10/genética , Camundongos , Camundongos Knockout , Neuralgia/genética , Neuralgia/imunologia , Neuralgia/patologia , Neurônios/patologia , Receptor A3 de Adenosina/genética
9.
PLoS One ; 15(12): e0243986, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33326493

RESUMO

Extracellular adenosine, a danger signal, can cause hypothermia. We generated mice lacking neuronal adenosine A1 receptors (A1AR, encoded by the Adora1 gene) to examine the contribution of these receptors to hypothermia. Intracerebroventricular injection of the selective A1AR agonist (Cl-ENBA, 5'-chloro-5'-deoxy-N6-endo-norbornyladenosine) produced hypothermia, which was reduced in mice with deletion of A1AR in neurons. A non-brain penetrant A1AR agonist [SPA, N6-(p-sulfophenyl) adenosine] also caused hypothermia, in wild type but not mice lacking neuronal A1AR, suggesting that peripheral neuronal A1AR can also cause hypothermia. Mice expressing Cre recombinase from the Adora1 locus were generated to investigate the role of specific cell populations in body temperature regulation. Chemogenetic activation of Adora1-Cre-expressing cells in the preoptic area did not change body temperature. In contrast, activation of Adora1-Cre-expressing dorsomedial hypothalamus cells increased core body temperature, concordant with agonism at the endogenous inhibitory A1AR causing hypothermia. These results suggest that A1AR agonism causes hypothermia via two distinct mechanisms: brain neuronal A1AR and A1AR on neurons outside the blood-brain barrier. The variety of mechanisms that adenosine can use to induce hypothermia underscores the importance of hypothermia in the mouse response to major metabolic stress or injury.


Assuntos
Hipotermia/metabolismo , Receptor A1 de Adenosina/metabolismo , Agonistas do Receptor A1 de Adenosina/farmacologia , Animais , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nervos Periféricos/metabolismo , Nervos Periféricos/fisiopatologia
10.
Cell Rep ; 31(2): 107501, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32294435

RESUMO

Human and mouse thermal physiology differ due to dissimilar body sizes. Unexpectedly, in mice we found no ambient temperature zone where both metabolic rate and body temperature were constant. Body temperature began increasing once cold-induced thermogenesis was no longer required. This result reproduced in male, female, C57BL/6J, 129, chow-fed, diet-induced obese, and ob/ob mice as well as Trpv1-/-;Trpm8-/-;Trpa1-/- mice lacking thermal sensory channels. During the resting-light phase, the energy expenditure minimum spanned ∼4°C of ambient temperature, whereas in the active-dark phase it approximated a point. We propose the concept of a thermoneutral point (TNP), a discrete ambient temperature below which energy expenditure increases and above which body temperature increases. Humans do not have a TNP. As studied, the mouse TNP is ∼29°C in light phase and ∼33°C in dark phase. These observations inform how thermoneutrality is defined and how mice are used to model human energy physiology and drug development.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Metabolismo Energético/fisiologia , Termogênese/fisiologia , Tecido Adiposo/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Composição Corporal/fisiologia , Tamanho Corporal/fisiologia , Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Temperatura Baixa , Ingestão de Energia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Temperatura
11.
Mol Metab ; 36: 100969, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32229422

RESUMO

OBJECTIVE: Bombesin-like receptor 3 (BRS3) is an orphan receptor and Brs3 knockout mice develop obesity with increased food intake and reduced resting metabolic rate and body temperature. The neuronal populations contributing to these effects were examined. METHODS: We studied energy metabolism in mice with Cre-mediated recombination causing 1) loss of BRS3 selectively in SIM1- or MC4R-expressing neurons or 2) selective re-expression of BRS3 from a null background in these neurons. RESULTS: The deletion of BRS3 in MC4R neurons increased body weight/adiposity, metabolic efficiency, and food intake, and reduced insulin sensitivity. BRS3 re-expression in these neurons caused partial or no reversal of these traits. However, these observations were confounded by an obesity phenotype caused by the Mc4r-Cre allele, independent of its recombinase activity. The deletion of BRS3 in SIM1 neurons increased body weight/adiposity and food intake, but not to the levels of the global null. The re-expression of BRS3 in SIM1 neurons reduced body weight/adiposity and food intake, but not to wild type levels. The deletion of BRS3 in either MC4R- or SIM1-expressing neurons affected body temperature, with re-expression in either population reversing the null phenotype. MK-5046, a BRS3 agonist, increases light phase body temperature in wild type, but not Brs3 null, mice and BRS3 re-expression in either population restored response to MK-5046. CONCLUSIONS: BRS3 in both MC4R- and SIM1-expressing neurons contributes to regulation of body weight/adiposity, insulin sensitivity, food intake, and body temperature.


Assuntos
Metabolismo Energético/fisiologia , Neurônios/metabolismo , Receptores da Bombesina/metabolismo , Adiposidade/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Temperatura Corporal/fisiologia , Peso Corporal , Encéfalo/metabolismo , Ingestão de Alimentos/fisiologia , Feminino , Homeostase/fisiologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Obesidade/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Receptores da Bombesina/genética , Proteínas Repressoras/metabolismo
12.
PLoS Biol ; 17(3): e3000161, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30822301

RESUMO

Adenosine is a constituent of many molecules of life; increased free extracellular adenosine indicates cell damage or metabolic stress. The importance of adenosine signaling in basal physiology, as opposed to adaptive responses to danger/damage situations, is unclear. We generated mice lacking all four adenosine receptors (ARs), Adora1-/-;Adora2a-/-;Adora2b-/-;Adora3-/- (quad knockout [QKO]), to enable investigation of the AR dependence of physiologic processes, focusing on body temperature. The QKO mice demonstrate that ARs are not required for growth, metabolism, breeding, and body temperature regulation (diurnal variation, response to stress, and torpor). However, the mice showed decreased survival starting at about 15 weeks of age. While adenosine agonists cause profound hypothermia via each AR, adenosine did not cause hypothermia (or bradycardia or hypotension) in QKO mice, indicating that AR-independent signals do not contribute to adenosine-induced hypothermia. The hypothermia elicited by adenosine kinase inhibition (with A134974), inosine, or uridine also required ARs, as each was abolished in the QKO mice. The proposed mechanism for uridine-induced hypothermia is inhibition of adenosine transport by uridine, increasing local extracellular adenosine levels. In contrast, adenosine 5'-monophosphate (AMP)-induced hypothermia was attenuated in QKO mice, demonstrating roles for both AR-dependent and AR-independent mechanisms in this process. The physiology of the QKO mice appears to be the sum of the individual knockout mice, without clear evidence for synergy, indicating that the actions of the four ARs are generally complementary. The phenotype of the QKO mice suggests that, while extracellular adenosine is a signal of stress, damage, and/or danger, it is less important for baseline regulation of body temperature.


Assuntos
Hipotermia/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptor A2B de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Animais , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Temperatura Corporal/genética , Temperatura Corporal/fisiologia , Cafeína/farmacologia , Feminino , Genótipo , Frequência Cardíaca/genética , Frequência Cardíaca/fisiologia , Hipotermia/induzido quimicamente , Hipotermia/genética , Inosina/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Receptor A1 de Adenosina/genética , Receptor A2A de Adenosina/genética , Receptor A2B de Adenosina/genética , Receptor A3 de Adenosina/genética , Uridina/toxicidade
13.
Nat Neurosci ; 21(11): 1530-1540, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30349101

RESUMO

Bombesin-like receptor 3 (BRS3) is an orphan G-protein-coupled receptor that regulates energy homeostasis and heart rate. We report that acute activation of Brs3-expressing neurons in the dorsomedial hypothalamus (DMHBrs3) increased body temperature (Tb), brown adipose tissue temperature, energy expenditure, heart rate, and blood pressure, with no effect on food intake or physical activity. Conversely, activation of Brs3 neurons in the paraventricular nucleus of the hypothalamus had no effect on Tb or energy expenditure, but suppressed food intake. Inhibition of DMHBrs3 neurons decreased Tb and energy expenditure, suggesting a necessary role in Tb regulation. We found that the preoptic area provides major input (excitatory and inhibitory) to DMHBrs3 neurons. Optogenetic stimulation of DMHBrs3 projections to the raphe pallidus increased Tb. Thus, DMHBrs3→raphe pallidus neurons regulate Tb, energy expenditure, and heart rate, and Brs3 neurons in the paraventricular nucleus of the hypothalamus regulate food intake. Brs3 expression is a useful marker for delineating energy metabolism regulatory circuitry.


Assuntos
Temperatura Corporal/fisiologia , Núcleo Hipotalâmico Dorsomedial/metabolismo , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Frequência Cardíaca/fisiologia , Neurônios/metabolismo , Receptores da Bombesina/metabolismo , Animais , Masculino , Camundongos
14.
Am J Physiol Endocrinol Metab ; 315(3): E357-E366, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29812984

RESUMO

Intraperitoneal administration of the melanocortin agonist melanotan II (MTII) to mice causes a profound, transient hypometabolism/hypothermia. It is preserved in mice lacking any one of melanocortin receptors 1, 3, 4, or 5, suggesting a mechanism independent of the canonical melanocortin receptors. Here we show that MTII-induced hypothermia was abolished in KitW-sh/W-sh mice, which lack mast cells, demonstrating that mast cells are required. MRGPRB2 is a receptor that detects many cationic molecules and activates mast cells in an antigen-independent manner. In vitro, MTII stimulated mast cells by both MRGPRB2-dependent and -independent mechanisms, and MTII-induced hypothermia was intact in MRGPRB2-null mice. Confirming that MTII activated mast cells, MTII treatment increased plasma histamine levels in both wild-type and MRGPRB2-null, but not in KitW-sh/W-sh, mice. The released histamine produced hypothermia via histamine H1 receptors because either a selective antagonist, pyrilamine, or ablation of H1 receptors greatly diminished the hypothermia. Other drugs, including compound 48/80, a commonly used mast cell activator, also produced hypothermia by both mast cell-dependent and -independent mechanisms. These results suggest that mast cell activation should be considered when investigating the mechanism of drug-induced hypothermia in mice.


Assuntos
Agonistas dos Receptores Histamínicos/farmacologia , Hipotermia/induzido quimicamente , Mastócitos/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , alfa-MSH/análogos & derivados , Animais , Liberação de Histamina/efeitos dos fármacos , Liberação de Histamina/genética , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , alfa-MSH/farmacologia
15.
Neuropharmacology ; 139: 268-278, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29548686

RESUMO

Extracellular adenosine is a danger/injury signal that initiates protective physiology, such as hypothermia. Adenosine has been shown to trigger hypothermia via agonism at A1 and A3 adenosine receptors (A1AR, A3AR). Here, we find that adenosine continues to elicit hypothermia in mice null for A1AR and A3AR and investigated the effect of agonism at A2AAR or A2BAR. The poorly brain penetrant A2AAR agonists CGS-21680 and PSB-0777 caused hypothermia, which was not seen in mice lacking A2AAR. MRS7352, a likely non-brain penetrant A2AAR antagonist, inhibited PSB-0777 hypothermia. While vasodilation is probably a contributory mechanism, A2AAR agonism also caused hypometabolism, indicating that vasodilation is not the sole mechanism. The A2BAR agonist BAY60-6583 elicited hypothermia, which was lost in mice null for A2BAR. Low intracerebroventricular doses of BAY60-6583 also caused hypothermia, indicating a brain site of action, with neuronal activation in the preoptic area and paraventricular nucleus of the hypothalamus. Thus, agonism at any one of the canonical adenosine receptors, A1AR, A2AAR, A2BAR, or A3AR, can cause hypothermia. This four-fold redundancy in adenosine-mediated initiation of hypothermia may reflect the centrality of hypothermia as a protective response.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Hipotermia/induzido quimicamente , Hipotermia/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptor A2B de Adenosina/metabolismo , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Receptor A2A de Adenosina/genética , Receptor A2B de Adenosina/genética
17.
Gastroenterology ; 154(1): 195-210, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28918914

RESUMO

BACKGROUND & AIMS: Development of hepatocellular carcinoma (HCC) is associated with alterations in the transforming growth factor-beta (TGF-ß) signaling pathway, which regulates liver inflammation and can have tumor suppressor or promoter activities. Little is known about the roles of specific members of this pathway at specific of HCC development. We took an integrated approach to identify and validate the effects of changes in this pathway in HCC and identify therapeutic targets. METHODS: We performed transcriptome analyses for a total of 488 HCCs that include data from The Cancer Genome Atlas. We also screened 301 HCCs reported in the Catalogue of Somatic Mutations in Cancer and 202 from Cancer Genome Atlas for mutations in genome sequences. We expressed mutant forms of spectrin beta, non-erythrocytic 1 (SPTBN1) in HepG2, SNU398, and SNU475 cells and measured phosphorylation, nuclear translocation, and transcriptional activity of SMAD family member 3 (SMAD3). RESULTS: We found somatic mutations in at least 1 gene whose product is a member of TGF-ß signaling pathway in 38% of HCC samples. SPTBN1 was mutated in the largest proportion of samples (12 of 202, 6%). Unsupervised clustering of transcriptome data identified a group of HCCs with activation of the TGF-ß signaling pathway (increased transcription of genes in the pathway) and a group of HCCs with inactivation of TGF-ß signaling (reduced expression of genes in this pathway). Patients with tumors with inactivation of TGF-ß signaling had shorter survival times than patients with tumors with activation of TGF-ß signaling (P = .0129). Patterns of TGF-ß signaling correlated with activation of the DNA damage response and sirtuin signaling pathways. HepG2, SNU398, and SNU475 cells that expressed the D1089Y mutant or with knockdown of SPTBN1 had increased sensitivity to DNA crosslinking agents and reduced survival compared with cells that expressed normal SPTBN1 (controls). CONCLUSIONS: In genome and transcriptome analyses of HCC samples, we found mutations in genes in the TGF-ß signaling pathway in almost 40% of samples. These correlated with changes in expression of genes in the pathways; up-regulation of genes in this pathway would contribute to inflammation and fibrosis, whereas down-regulation would indicate loss of TGF-ß tumor suppressor activity. Our findings indicate that therapeutic agents for HCCs can be effective, based on genetic features of the TGF-ß pathway; agents that block TGF-ß should be used only in patients with specific types of HCCs.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Mutação/genética , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Idoso , Carcinoma Hepatocelular/mortalidade , Estudos de Casos e Controles , Análise por Conglomerados , Feminino , Humanos , Neoplasias Hepáticas/mortalidade , Masculino , Pessoa de Meia-Idade
18.
Mol Metab ; 6(11): 1540-1550, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29107299

RESUMO

OBJECTIVE: Bombesin-like receptor 3 (BRS-3) is an orphan G protein-coupled receptor. Brs3 null mice have reduced resting metabolic rate and body temperature, increased food intake, and obesity. Here we study the role of Brs3 in different neuron types. METHODS: Mice able to undergo Cre recombinase-dependent inactivation or re-expression of Brs3 were generated, respectively Brs3fl/y and Brs3loxTB/y. We then studied four groups of mice with Brs3 selectively inactivated or re-expressed in cells expressing Vglut2-Cre or Vgat-Cre. RESULTS: Deletion of Brs3 in glutamatergic neurons expressing Vglut2 reproduced the global null phenotype for regulation of food intake, metabolic rate, body temperature, adiposity, and insulin resistance. These mice also no longer responded to a BRS-3 agonist, MK-5046. In contrast, deletion of Brs3 in GABAergic neurons produced no detectable phenotype. Conversely, the wild type phenotype was restored by selective re-expression of Brs3 in glutamatergic neurons, with no normalization achieved by re-expressing Brs3 in GABAergic neurons. CONCLUSIONS: Brs3 expression in glutamatergic neurons is both necessary and sufficient for full Brs3 function in energy metabolism. In these experiments, no function was identified for Brs3 in GABAergic neurons. The data suggest that the anti-obesity pharmacologic actions of BRS-3 agonists occur via agonism of receptors on glutamatergic neurons.


Assuntos
Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Receptores da Bombesina/biossíntese , Adiposidade , Animais , Peso Corporal , Ingestão de Alimentos/fisiologia , Metabolismo Energético , Neurônios GABAérgicos/metabolismo , Expressão Gênica , Imidazóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Obesidade/metabolismo , Pirazóis/farmacologia , Receptores da Bombesina/agonistas , Receptores da Bombesina/genética
19.
Cell Rep ; 18(13): 3155-3166, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28355567

RESUMO

Preadipocytes initiate differentiation into adipocytes through a cascade of events. Mitotic clonal expansion, as one of the earliest events, is essential for adipogenesis. However, the underlying mechanisms that regulate mitotic clonal expansion remain elusive. SIRT6 is a member of the evolutionarily conserved sirtuin family of nicotinamide adenine dinucleotide (NAD)+-dependent protein deacetylases. Here, we show that SIRT6 deficiency in preadipocytes blocks their adipogenesis. Analysis of gene expression during adipogenesis reveals that KIF5C, which belongs to the kinesin family, is negatively regulated by SIRT6. Furthermore, we show that KIF5C is a negative factor for adipogenesis through interacting with CK2α', a catalytic subunit of CK2. This interaction blocks CK2α' nuclear translocation and CK2 kinase activity and inhibits mitotic clonal expansion during adipogenesis. These findings reveal a crucial role of SIRT6 in adipogenesis and provide potential therapeutic targets for obesity.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Diferenciação Celular , Mitose , Sirtuínas/metabolismo , Células 3T3-L1 , Adipogenia , Animais , Caseína Quinase II/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células , Células Clonais , Deleção de Genes , Cinesinas/metabolismo , Camundongos , Fenótipo , Transporte Proteico , Sirtuínas/deficiência
20.
Neuropharmacology ; 114: 101-113, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27914963

RESUMO

Small mammals have the ability to enter torpor, a hypothermic, hypometabolic state, allowing impressive energy conservation. Administration of adenosine or adenosine 5'-monophosphate (AMP) can trigger a hypothermic, torpor-like state. We investigated the mechanisms for hypothermia using telemetric monitoring of body temperature in wild type and receptor knock out (Adora1-/-, Adora3-/-) mice. Confirming prior data, stimulation of the A3 adenosine receptor (AR) induced hypothermia via peripheral mast cell degranulation, histamine release, and activation of central histamine H1 receptors. In contrast, A1AR agonists and AMP both acted centrally to cause hypothermia. Commonly used, selective A1AR agonists, including N6-cyclopentyladenosine (CPA), N6-cyclohexyladenosine (CHA), and MRS5474, caused hypothermia via both A1AR and A3AR when given intraperitoneally. Intracerebroventricular dosing, low peripheral doses of Cl-ENBA [(±)-5'-chloro-5'-deoxy-N6-endo-norbornyladenosine], or using Adora3-/- mice allowed selective stimulation of A1AR. AMP-stimulated hypothermia can occur independently of A1AR, A3AR, and mast cells. A1AR and A3AR agonists and AMP cause regulated hypothermia that was characterized by a drop in total energy expenditure, physical inactivity, and preference for cooler environmental temperatures, indicating a reduced body temperature set point. Neither A1AR nor A3AR was required for fasting-induced torpor. A1AR and A3AR agonists and AMP trigger regulated hypothermia via three distinct mechanisms.


Assuntos
Agonistas do Receptor A1 de Adenosina/administração & dosagem , Agonistas do Receptor A3 de Adenosina/administração & dosagem , Monofosfato de Adenosina/fisiologia , Febre/induzido quimicamente , Receptor A1 de Adenosina/fisiologia , Receptor A3 de Adenosina/fisiologia , Torpor , Adenosina/administração & dosagem , Adenosina/análogos & derivados , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Histamina/metabolismo , Injeções Intraventriculares , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptor A1 de Adenosina/genética , Receptor A3 de Adenosina/genética , Receptores Histamínicos H1/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA