Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Nat Commun ; 11(1): 6378, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33311503

RESUMEN

Homeotherms maintain a stable internal body temperature despite changing environments. During energy deficiency, some species can cease to defend their body temperature and enter a hypothermic and hypometabolic state known as torpor. Recent advances have revealed the medial preoptic area (MPA) as a key site for the regulation of torpor in mice. The MPA is estrogen-sensitive and estrogens also have potent effects on both temperature and metabolism. Here, we demonstrate that estrogen-sensitive neurons in the MPA can coordinate hypothermia and hypometabolism in mice. Selectively activating estrogen-sensitive MPA neurons was sufficient to drive a coordinated depression of metabolic rate and body temperature similar to torpor, as measured by body temperature, physical activity, indirect calorimetry, heart rate, and brain activity. Inducing torpor with a prolonged fast revealed larger and more variable calcium transients from estrogen-sensitive MPA neurons during bouts of hypothermia. Finally, whereas selective ablation of estrogen-sensitive MPA neurons demonstrated that these neurons are required for the full expression of fasting-induced torpor in both female and male mice, their effects on thermoregulation and torpor bout initiation exhibit differences across sex. Together, these findings suggest a role for estrogen-sensitive MPA neurons in directing the thermoregulatory and metabolic responses to energy deficiency.


Asunto(s)
Temperatura Corporal/fisiología , Estrógenos/metabolismo , Neuronas/fisiología , Área Preóptica/metabolismo , Letargo/fisiología , Animales , Temperatura Corporal/genética , Regulación de la Temperatura Corporal/fisiología , Metabolismo Energético/fisiología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Ayuno , Femenino , Hipotermia/genética , Hipotermia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
J Pharmacol Sci ; 144(2): 61-68, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32684333

RESUMEN

The effects of adipokine administration to the hypothalamic preoptic area (POA), which is one of the body temperature (BT) regulation centers in the central nervous system, on BT were investigated in male Wistar rats. BT was measured in conscious rats using telemetry. Insulin-like growth factor-1 (IGF-1), interleukin-1ß (IL-1ß), monocyte chemoattractant protein-1 and lipocalin-2 produced hyperthermia, and the effects induced by IL-1ß (25 ng) and IGF-1 (5 µg) were sustainable and remarkable. IL-6 did not show any significant effect. The IGF-1-induced effect was inhibited by pretreatment with IGF binding protein 3 (IGFBP3) or NVP-AEW541 (NVP, a selective inhibitor of type 1 IGF receptor tyrosine kinase, IGF1R TK). NVP-induced inhibition was observed only in the early phase of IGF-1-induced hyperthermia. In addition, IGF-1 increased the IL-1ß concentration in the microdialysate of POA perfusion, but did not increase the IL-1ß concentration in the plasma or the PGE2 concentration in the microdialysate. These findings suggested that IGF-1 produced hyperthermia, which was mediated, at least a part, through an increased IL-1ß concentration after activation of IGF1R TK in the POA, and the IGF-IGFBP system possibly participates in BT homeostasis in the POA.


Asunto(s)
Adipoquinas/administración & dosificación , Adipoquinas/farmacología , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/genética , Área Preóptica/metabolismo , Área Preóptica/fisiología , Animales , Quimiocina CCL2/administración & dosificación , Quimiocina CCL2/farmacología , Fiebre/inducido químicamente , Fiebre/genética , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/farmacología , Interleucina-1beta/administración & dosificación , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Lipocalina 2/administración & dosificación , Lipocalina 2/farmacología , Masculino , Proteínas Tirosina Quinasas/metabolismo , Ratas Wistar , Receptor IGF Tipo 1/metabolismo
3.
FASEB J ; 34(1): 107-121, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914628

RESUMEN

The peptide kisspeptin and its receptor, Kiss1r, act centrally to stimulate reproduction. Evidence indicates that kisspeptin signaling is also important for body weight (BW) and metabolism. We recently reported that Kiss1r KO mice develop obesity, along with reduced metabolism and energy expenditure, independent of estradiol levels. Outside the brain, Kiss1r is expressed in several metabolic tissues, including brown adipose tissue (BAT), but it is unknown which specific tissue is responsible for the metabolic phenotype in Kiss1r KOs. We first determined that global Kiss1r KO mice have significant alterations in body temperature and BAT thermogenic gene expression, perhaps contributing to their obesity. Next, to test whether kisspeptin signaling specifically in BAT influences BW, metabolism, or body temperature, we used Cre/lox technology to generate conditional Kiss1r knockout exclusively in BAT (BAT-Kiss1r KO). Unlike global Kiss1r KOs, BAT-Kiss1r KOs (lacking Kiss1r in just BAT) were not hypogonadal, as expected. Surprisingly, however, BAT-Kiss1r KOs of both sexes displayed significantly lower BW and adiposity than controls. This novel BAT-Kiss1r KO phenotype was of greater magnitude in females and was associated with improved glucose tolerance, increased metabolism, energy expenditure, and locomotor activity, along with increased body temperature and BAT gene expression, specifically Cox8b. Our findings suggest that the previously observed obesity and decreased metabolism in global Kiss1r KOs reflect impaired kisspeptin signaling in non-BAT tissues. However, the novel finding of increased metabolism and body temperature and lower BW in BAT-Kiss1r KOs reveal a previously unidentified role for endogenous kisspeptin signaling in BAT in modulating metabolic and thermogenic physiology.


Asunto(s)
Adipocitos Marrones/metabolismo , Temperatura Corporal/fisiología , Peso Corporal/fisiología , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Receptores de Kisspeptina-1/metabolismo , Animales , Temperatura Corporal/genética , Peso Corporal/genética , Genotipo , Ratones , Ratones Noqueados , Receptores de Kisspeptina-1/genética
4.
J Biol Chem ; 295(7): 2034-2042, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31919095

RESUMEN

Prolonged cold exposure stimulates the formation of brownlike adipocytes expressing UCP1 (uncoupling-protein-1) in subcutaneous white adipose tissue which, together with classical brown adipose tissue, contributes to maintaining body temperature in mammals through nonshivering thermogenesis. The mechanisms that regulate the formation of these cells, alternatively called beige or brite adipocytes, are incompletely understood. Here we report that mice lacking CD137, a cell surface protein used in several studies as a marker for beige adipocytes, showed elevated levels of thermogenic markers, including UCP1, increased numbers of beige adipocyte precursors, and expanded UCP1-expressing cell clusters in inguinal white adipose tissue after chronic cold exposure. CD137 knockout mice also showed enhanced cold resistance. These results indicate that CD137 functions as a negative regulator of "browning" in white adipose tissue and call into question the use of this protein as a functional marker for beige adipocytes.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Proteína Desacopladora 1/genética , Adipocitos Beige/metabolismo , Animales , Temperatura Corporal/genética , Frío , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Noqueados , Termogénesis/genética
5.
Diabetologia ; 62(12): 2325-2339, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31511929

RESUMEN

AIMS/HYPOTHESIS: Absent in melanoma 2 (AIM2) is a cytosolic sensor for double-stranded DNA and a tumour suppressor. Binding of double-stranded DNA to AIM2 forms the AIM2 inflammasome, leading to activation of caspase-1 and production of IL-1ß and IL-18. Although inflammasome-independent effects of AIM2 have been reported, its role in energy metabolism is unknown. We aimed to evaluate the effect of AIM2 in energy metabolism and glucose homeostasis. METHODS: Male and female whole body Aim2 knockout (Aim2-/-) mice were used in the current study. Body weight, food intake, body composition, energy expenditure, fasting blood glucose levels, GTT and body temperature were measured at indicated time points. RNA sequencing was carried out on gonadal white adipose tissue (gWAT) in 14-month-old female mice. mRNA and protein levels in tissues were analysed by quantitative real-time PCR and immunoblot. Immune cell infiltration in gWAT was examined by flow cytometry. Stromal vascular fractions isolated from gWAT were used to investigate adipocyte differentiation. RESULTS: Male and female Aim2-/- mice were obese compared with wild-type controls from 7 weeks of age until 51 weeks of age, with increased adiposity in both subcutaneous and visceral fat depots. While there were no differences in food intake, Aim2-/- mice demonstrated decreased energy expenditure and impaired brown adipose tissue function compared with wild-type controls. Fasting glucose and insulin levels were elevated, and Aim2-/- mice were glucose intolerant on intraperitoneal GTT. RNA sequencing revealed marked upregulation of the IFN-inducible gene Ifi202b, which encodes protein 202 (p202) and elevated inflammatory signalling in gWAT of Aim2-/- mice. Increased infiltration of total and Ly6Clow monocytes was noted at 8 weeks of age in gWAT, before the onset of obesity and insulin resistance. Ifi202b knockdown blocked adipogenesis in stromal vascular fractions and reduced inflammation in bone marrow-derived macrophages, demonstrating a key role of p202 in mediating the increased adipogenesis and inflammation in Aim2-/- mice. CONCLUSIONS/INTERPRETATION: These results demonstrate a fundamental role for AIM2 in energy metabolism, inflammation and insulin resistance. Our studies establish a novel link between the innate immunity proteins, AIM2 and p202, and metabolism.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo Blanco/metabolismo , Proteínas de Unión al ADN/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina/genética , Obesidad/metabolismo , Tejido Adiposo Pardo/metabolismo , Adiposidad/genética , Animales , Glucemia/metabolismo , Temperatura Corporal/genética , Proteínas de Unión al ADN/genética , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Ayuno/metabolismo , Femenino , Inflamación/genética , Masculino , Ratones , Ratones Noqueados , Obesidad/genética
6.
Brain Behav Immun ; 81: 260-271, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31220563

RESUMEN

Increased production of pro-inflammatory cytokines is assumed to mediate increased sleep under inflammatory conditions, such as systemic infections or recovery from sleep loss. The role of cytokines in sleep regulation under normal conditions is less clear. In the present study, we investigated the role of endogenous tumor necrosis factor alpha (TNFα) in sleep regulation using TNFα knockout (KO) mice. Under control conditions at thermoneutral ambient temperature, total sleep time did not differ between TNFα KO and wild-type (WT) mice, but TNFα KO mice had increased rapid-eye-movement sleep (REMS), accompanied by decreased motor activity and body temperature. Exposure to 17 °C induced decreases in total sleep time similarly in both genotypes. Sleep deprivation by gentle handling elicited robust rebound increases in non-rapid-eye movement sleep (NREMS), REMS and electroencephalographic (EEG) slow-wave activity (SWA), accompanied by suppressed motor activity and decreased body temperature; there was no significant difference between the responses of WT and KO mice. Systemic injection of the beta3-adrenergic receptor (ß3-AR) agonist CL-316,243 induced increases in NREMS and body temperature. The temperature response, but not the sleep effect, was attenuated in the KO animals. Systemic injection of TNFα induced increased NREMS, reduced REMS and biphasic temperature responses in both genotypes. In the KO mice, the NREMS-promoting effects of exogenously administered TNFα was decreased, while REMS suppression was enhanced, and the first, hypothermic, phase of temperature response was attenuated. Overall, TNFα KO mice did not show any deficiency in sleep regulation which suggests that the role of endogenous TNFα in sleep regulation is less pronounced than previously suggested.


Asunto(s)
Temperatura Corporal/fisiología , Sueño/fisiología , Factor de Necrosis Tumoral alfa/genética , Animales , Temperatura Corporal/genética , Dioxoles/farmacología , Electroencefalografía , Hipotermia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/genética , Actividad Motora/fisiología , Polisomnografía , Receptores Adrenérgicos beta 3/metabolismo , Sueño/genética , Privación de Sueño/genética , Privación de Sueño/metabolismo , Fases del Sueño , Sueño REM/genética , Sueño REM/fisiología , Temperatura , Factor de Necrosis Tumoral alfa/metabolismo , Vigilia/fisiología
7.
Am J Physiol Regul Integr Comp Physiol ; 317(2): R240-R247, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31188649

RESUMEN

Cold-shock proteins are thought to participate in the cold-tolerant nature of hibernating animals. We previously demonstrated that an alternative splicing may allow rapid induction of functional cold-inducible RNA-binding protein (CIRBP) in the hamster heart. The purpose of the present study was to determine the major cause of the alternative splicing in Syrian hamsters. RT-PCR analysis revealed that CIRBP mRNA is constitutively expressed in the heart, brain, lung, liver, and kidney of nonhibernating euthermic hamsters with several alternative splicing variants. In contrast, the short variant containing an open-reading frame for functional CIRBP was dominantly found in the hibernating animals. Keeping the animals in a cold and dark environment did not cause a shift in the alternative splicing. Induction of hypothermia by central administration of an adenosine A1-receptor agonist reproduced the shift in the splicing pattern. However, the agonist failed to shift the pattern when body temperature was kept at 37°C, suggesting that central adenosine A1 receptors are not directly linked to the shift of the alternative splicing. Rapid reduction of body temperature to 10°C by isoflurane anesthesia combined with cooling did not alter the splicing pattern, but maintenance of mild hypothermia (~28°C) for 2 h elicited the shift in the pattern. The results suggest that animals need to be maintained at mild hypothermia for an adequate duration to induce the shift in the alternative splicing. This is applicable to natural hibernation because hamsters entering hibernation show a gradual decrease in body temperature, being maintained at mild hypothermia for several hours.


Asunto(s)
Empalme Alternativo/genética , Frío , Hibernación/genética , Hipotermia/fisiopatología , Proteínas de Unión al ARN/metabolismo , Aclimatación/fisiología , Animales , Temperatura Corporal/genética , Temperatura Corporal/fisiología , Corazón/fisiología , Hibernación/fisiología , Masculino , ARN Mensajero/metabolismo
8.
Diabetes ; 68(6): 1130-1142, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30862681

RESUMEN

Obesity and related inflammation are critical for the pathogenesis of insulin resistance, but the underlying mechanisms are not fully understood. Formyl peptide receptor 2 (FPR2) plays important roles in host immune responses and inflammation-related diseases. We found that Fpr2 expression was elevated in the white adipose tissue of high-fat diet (HFD)-induced obese mice and db/db mice. The systemic deletion of Fpr2 alleviated HFD-induced obesity, insulin resistance, hyperglycemia, hyperlipidemia, and hepatic steatosis. Furthermore, Fpr2 deletion in HFD-fed mice elevated body temperature, reduced fat mass, and inhibited inflammation by reducing macrophage infiltration and M1 polarization in metabolic tissues. Bone marrow transplantations between wild-type and Fpr2-/- mice and myeloid-specific Fpr2 deletion demonstrated that Fpr2-expressing myeloid cells exacerbated HFD-induced obesity, insulin resistance, glucose/lipid metabolic disturbances, and inflammation. Mechanistic studies revealed that Fpr2 deletion in HFD-fed mice enhanced energy expenditure probably through increasing thermogenesis in skeletal muscle; serum amyloid A3 and other factors secreted by adipocytes induced macrophage chemotaxis via Fpr2; and Fpr2 deletion suppressed macrophage chemotaxis and lipopolysaccharide-, palmitate-, and interferon-γ-induced macrophage M1 polarization through blocking their signals. Altogether, our studies demonstrate that myeloid Fpr2 plays critical roles in obesity and related metabolic disorders via regulating muscle energy expenditure, macrophage chemotaxis, and M1 polarization.


Asunto(s)
Quimiotaxis/genética , Dieta Alta en Grasa , Resistencia a la Insulina/genética , Macrófagos/inmunología , Receptores de Formil Péptido/genética , Animales , Temperatura Corporal/genética , Metabolismo Energético/genética , Hígado Graso/genética , Hígado Graso/inmunología , Hiperglucemia/genética , Hiperglucemia/inmunología , Hiperlipidemias/genética , Hiperlipidemias/inmunología , Inflamación/genética , Inflamación/inmunología , Resistencia a la Insulina/inmunología , Ratones , Ratones Noqueados , Ratones Obesos , Proteína Amiloide A Sérica/metabolismo , Termogénesis/genética
9.
PLoS Biol ; 17(3): e3000161, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30822301

RESUMEN

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.


Asunto(s)
Hipotermia/metabolismo , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2B/metabolismo , Receptor de Adenosina A3/metabolismo , Animales , Presión Sanguínea/genética , Presión Sanguínea/fisiología , Temperatura Corporal/genética , Temperatura Corporal/fisiología , Cafeína/farmacología , Femenino , Genotipo , Frecuencia Cardíaca/genética , Frecuencia Cardíaca/fisiología , Hipotermia/inducido químicamente , Hipotermia/genética , Inosina/farmacología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Receptor de Adenosina A1/genética , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2B/genética , Receptor de Adenosina A3/genética , Uridina/toxicidad
10.
Epilepsy Res ; 134: 1-8, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28505490

RESUMEN

It has been established that febrile seizures and its extended syndromes like generalized epilepsy with febrile seizures (FS) plus (GEFS+) and Dravet syndrome have been associated with mutations especially in SCN1A and GABRG2 genes. In patients, the onset of FS is likely due to the combined effect of temperature and inflammation in genetically vulnerable individuals because fever is often associated with infection. Much effort has been spent to understand the mechanisms underlying fever induction of seizures. In addition to the role of cytokines in FS, previous studies in Scn1a+/- knockout mice, a model of Dravet syndrome, indicated that temperature elevation alone could result in seizure generation, and the effect of elevated temperature inducing seizures was age-dependent. Here, we report the thermal effect in a different mouse model of Dravet syndrome, the Gabrg2+/Q390X knockin mouse. We demonstrated age-dependent dysregulated temperature control and that temperature elevation produced myoclonic jerks, generalized tonic clonic seizures (GTCSs) and heightened anxiety-like symptoms in Gabrg2+/Q390X mice. The study indicated that regardless of other inflammatory factors, brief heat alone increased brain excitability and induced multiple types of seizures in Gabrg2+/Q390X mice, suggesting that mutations like GABRG2(Q390X) may alter brain thermal regulation and precipitate seizures during temperature elevations.


Asunto(s)
Epilepsias Mioclónicas/complicaciones , Epilepsias Mioclónicas/genética , Calor/efectos adversos , Receptores de GABA-A/genética , Convulsiones/etiología , Factores de Edad , Animales , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/genética , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsias Mioclónicas/etiología , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Canal de Sodio Activado por Voltaje NAV1.1/deficiencia , Canal de Sodio Activado por Voltaje NAV1.1/genética , Pentilenotetrazol/toxicidad , Convulsiones/genética , Factor de Necrosis Tumoral alfa
11.
Sleep ; 40(6)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28444394

RESUMEN

Study objectives: Shortened or mistimed sleep affects metabolic homeostasis, which may in part be mediated by dysregulation of endogenous circadian clocks. In this study, we assessed the contribution of sleep disruption to metabolic dysregulation by analysing diurnal transcriptome regulation in metabolic tissues of mice subjected to a sleep restriction (SR) paradigm. Methods: Male mice were subjected to 2 × 5 days of SR with enforced waking during the first 6 hours of the light phase. SR and control mice were sacrificed at different time points of the day and RNA preparations from the mediobasal hypothalamus (MBH), liver, and epididymal white adipose tissue (eWAT) were subjected to whole-genome microarray hybridization. Transcriptional rhythms were associated with changes in behavioral and physiological parameters such as sleep, body temperature, and food intake. Rhythm detection was performed with CircWave and transcription profiles were compared by 2-way analysis of variance and t-tests with Benjamini-Hochberg corrections. Results: Clock gene rhythms were blunted in all tissues, while transcriptome regulation was associated with either clock gene expression, sleep patterns, or food intake in a tissue-specific manner. Clock gene expression was associated with apoptosis pathways in the MBH and with tumor necrosis factor alpha signalling in liver. Food intake-associated genes included cilium movement genes in the MBH and lipid metabolism-associated transcripts in liver. Conclusions: In mice, repeated SR profoundly alters behavioral and molecular diurnal rhythms, disrupting essential signalling pathways in MBH, liver, and eWAT, which may underlie the metabolic and cognitive disturbances observed in sleep-restricted humans such as shift workers.


Asunto(s)
Ritmo Circadiano/genética , Especificidad de Órganos/genética , Privación de Sueño/genética , Transcriptoma , Tejido Adiposo Blanco/metabolismo , Animales , Apoptosis/genética , Temperatura Corporal/genética , Relojes Circadianos/genética , Ingestión de Alimentos/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Masculino , Ratones , Sueño/genética , Factor de Necrosis Tumoral alfa/metabolismo
12.
Pain ; 157(11): 2561-2570, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27437788

RESUMEN

Cold exposure and a variety of types of mild stress increase pain in patients with painful disorders such as fibromyalgia syndrome. Acutely, stress induces thermogenesis by increasing sympathetic activation of beta-3 (ß3) adrenergic receptors in brown adipose tissue. Chronic stress leads to the hypertrophy of brown adipose, a phenomenon termed adaptive thermogenesis. Based on the innervation of skeletal muscle by collaterals of nerves projecting to brown adipose, we theorized an association between brown adipose tissue activity and musculoskeletal hyperalgesia and tested this hypothesis in mice. Exposure to a cold swim or injection of BRL37344 (ß3 adrenergic agonist) each enhanced musculoskeletal hyperalgesia, as indicated by morphine-sensitive decreases in grip force responses, whereas SR59230A (ß3 adrenergic antagonist) attenuated swim-induced hyperalgesia. Chemical ablation of interscapular brown adipose, using Rose Bengal, attenuated the development of hyperalgesia in response to either swim stress or BRL37344. In addition, elimination of the gene expressing uncoupling protein-1 (UCP1), the enzyme responsible for thermogenesis, prevented musculoskeletal hyperalgesia in response to either a swim or BRL37344, as documented in UCP1-knockout (UCP1-KO) mice compared with wild-type controls. Together, these data provide a convergence of evidence suggesting that activation of brown adipose contributes to stress-induced musculoskeletal hyperalgesia.


Asunto(s)
Tejido Adiposo Pardo/patología , Hiperalgesia/etiología , Hiperalgesia/patología , Dolor Musculoesquelético/complicaciones , Tejido Adiposo Pardo/efectos de los fármacos , Agonistas Adrenérgicos beta/toxicidad , Animales , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/genética , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Frío/efectos adversos , Modelos Animales de Enfermedad , Etanolaminas/toxicidad , Femenino , Hiperalgesia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fuerza Muscular/efectos de los fármacos , Dolor Musculoesquelético/patología , Dolor Musculoesquelético/cirugía , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Natación/psicología , Cola (estructura animal)/inervación , Proteína Desacopladora 1/deficiencia , Proteína Desacopladora 1/genética
13.
CNS Neurol Disord Drug Targets ; 14(5): 612-26, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25921737

RESUMEN

Transmembrane AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor regulatory protein (TARP) γ-8 is an auxiliary protein associated with some AMPA receptors. Most strikingly, AMPA receptors associated with this TARP have a relatively high localization in the hippocampus. TARP γ-8 also modifies the pharmacology and trafficking of AMPA receptors. However, to date there is little understanding of the biological significance of this auxiliary protein. In the present set of studies we provide a characterization of the differential pharmacology and behavioral consequences of deletion of TARP γ-8 by comparing the wild type (WT) and γ-8 -/- (knock-out, KO) mouse. KO mice were mildly hyperactive in a locomotor arena but not in other environments compared to WT mice. Additionally, the KO mice demonstrated enhanced locomotor stimulatory effects of both d-amphetamine and phencyclidine. Marble-burying and digging behaviors were dramatically reduced in KO mice. In another assay that can detect anxiety-like phenotypes, the elevated plus maze, no differences were observed in overall movement or open arm entries. In the forced-swim assay, KO mice displayed decreases in immobility time like the antidepressant imipramine and the AMPA receptor potentiator, LY392098. In KO mice, the antidepressant-like effects of LY392098 were prevented whereas the effects of imipramine were unaffected. Convulsions were induced by pentylenetetrazole, N-methyl-D-aspartate, and by kainic acid. However, in KO mice, kainic acid produced less tonic convulsions and lethality. KO mice had reduced levels of norepinephrine in hippocampus and cerebellum but not in hypothalamus or prefrontal cortex, decreased levels of cAMP in hippocampus, and increased levels of acetylcholine in the hypothalamus and prefrontal cortex. KO mice displayed decreased turnover of dopamine and increased histamine turnover in multiple brain areas In contrast, serotonin and its metabolites were not significantly affected by deletion of the γ-8 protein. Of a large panel of plasma lipids, only two monoacylglycerols (1OG and 2OG) were marginally but nonsignificantly altered in WT vs KO mice. Overall, the data suggest genetic inactivation of this specific population of AMPA receptors results in modest changes in behavior characterized by a mild hyperactivity which is condition dependent and a marked reduction in digging and burying behaviors. Despite deletion of TARP γ-8, chemoconvulsants were still active. Consistent with their predicted pharmacological actions, the convulsant effects of kainate and the antidepressant-like effects of an AMPA receptor potentiator (both acting upon AMPA receptors) were reduced or absent in KO mice.


Asunto(s)
Temperatura Corporal/genética , Encéfalo/metabolismo , Canales de Calcio/deficiencia , Hipercinesia/genética , Actividad Motora/genética , Receptores AMPA/metabolismo , Acetilcolina/metabolismo , Anfetaminas/farmacología , Animales , Monoaminas Biogénicas/metabolismo , Canales de Calcio/genética , Estimulantes del Sistema Nervioso Central/farmacología , AMP Cíclico/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Conducta Exploratoria/fisiología , Histamina/metabolismo , Lípidos/sangre , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Pentilenotetrazol , Fenciclidina/farmacología , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Convulsiones/genética , Sulfonamidas/farmacología , Natación/psicología , Tiofenos/farmacología , Factores de Tiempo
14.
Neuropsychopharmacology ; 40(9): 2175-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25740289

RESUMEN

Continued methamphetamine (MA) use is dependent on a positive MA experience and is likely attenuated by sensitivity to the aversive effects of MA. Bidirectional selective breeding of mice for high (MAHDR) or low (MALDR) voluntary consumption of MA demonstrates a genetic influence on MA intake. Quantitative trait locus (QTL) mapping identified a QTL on mouse chromosome 10 that accounts for greater than 50% of the genetically-determined differences in MA intake in the MAHDR and MALDR lines. The trace amine-associated receptor 1 gene (Taar1) is within the confidence interval of the QTL and encodes a receptor (TAAR1) that modulates monoamine neurotransmission and at which MA serves as an agonist. We demonstrate the existence of a non-functional allele of Taar1 in the DBA/2J mouse strain, one of the founder strains of the selected lines, and show that this non-functional allele co-segregates with high MA drinking and with reduced sensitivity to MA-induced conditioned taste aversion (CTA) and hypothermia. The functional Taar1 allele, derived from the other founder strain, C57BL/6J, segregates with low MA drinking and heightened sensitivity to MA-induced CTA and hypothermia. A role for TAAR1 in these phenotypes is corroborated in Taar1 transgenic mice: Taar1 knockout mice consume more MA and exhibit insensitivity to MA-induced CTA and hypothermia, compared with Taar1 wild-type mice. These are the first data to show that voluntary MA consumption is, in part, regulated by TAAR1 function. Behavioral and physiological studies indicate that TAAR1 function increases sensitivity to aversive effects of MA, and may thereby protect against MA use.


Asunto(s)
Estimulantes del Sistema Nervioso Central/administración & dosificación , Conducta de Elección/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Metanfetamina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/genética , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Metanfetamina/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Modelos Moleculares , Mutación/genética , Receptores Acoplados a Proteínas G/genética , Gusto/efectos de los fármacos , Gusto/genética , Transfección
15.
Growth Horm IGF Res ; 25(2): 80-4, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25588992

RESUMEN

OBJECTIVE: Growth hormone (GH) deficiency (GHD) leads to growth failure and changes in body composition, including increased fat accumulation and reduced lean body mass in both humans and rodents. The aim of this study was to examine the factors that contribute to energy imbalance in the GH releasing hormone knock out (GHRHKO) mice, a well established model of GHD. DESIGN: We evaluated food intake (of standard laboratory chow), total body weight (TBW), locomotor activity, body temperature and interscapular brown adipose tissue (BAT) weight in 8 adult male mice homozygous for the GHRHKO allele (-/-) and 8 heterozygous (+/-) animals as controls. The gene expression of uncoupling protein-1 (UCP-1) in BAT and the levels of norepinephrine (NE), dopamine (DA), and serotonin (5-hydroxytryptamine, 5-HT) in the ventral striatum were measured by real-time reverse transcription polymerase chain reaction (RT-PCR) and high performance liquid chromatography (HPLC) analysis, respectively. RESULTS: Throughout 2 months of observation -/- mice consumed approximately 40% more food (normalized to TBW; P<0.001), and showed increased locomotor activity in 24h time compared to controls (P<0.05). Moreover, -/- animals showed increased body temperature (P<0.001), BAT weight (P<0.001), and UCP-1 gene expression (P<0.001), while NE levels in the striatum area were lower (P<0.05) than controls. CONCLUSIONS: The present study demonstrates that the increased food intake observed in GHRH ablated animals is associated with increased locomotor and thermogenic activity.


Asunto(s)
Hormona Liberadora de Hormona del Crecimiento/genética , Actividad Motora/genética , Termogénesis/genética , Tejido Adiposo Pardo/anatomía & histología , Tejido Adiposo Pardo/metabolismo , Animales , Temperatura Corporal/genética , Ingestión de Alimentos/genética , Femenino , Eliminación de Gen , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Tamaño de los Órganos/genética , Proteína Desacopladora 1
16.
Anesthesiology ; 121(6): 1258-69, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25089642

RESUMEN

BACKGROUND: Myeloid differentiation factor 88 (MyD88) is an adaptor molecule critical for host innate immunity. Studies have shown that signaling via MyD88 contributes to cytokine storm, cardiac dysfunction, and high mortality during endotoxin shock.However, the specific contribution of MyD88 signaling of immune and cardiac origins to endotoxin shock remains unknown. METHODS: Tissue-specific MyD88 deletion models: Cre-recombinase transgenic mice with α-myosin heavy chain (α-MHC) or lysozyme M promoters were cross-bred with MyD88-loxP (MyD88fl/fl) mice, respectively, to generate cardiomyocyte- (α-MHCMyD88−/−) or myeloid-specific (Lyz-MyD88−/−) MyD88 deletion models and their respective MyD88fl/fl littermates. Endotoxin shock model: Mice were subjected to 15 mg/kg lipopolysaccharide (intraperitoneal injection). Cardiac function was measured by echocardiography and cytokines by multiplex assay and quantitative reverse transcription-polymerase chain reaction. RESULTS: α-MHC-MyD88−/− mice had 61 and 87% reduction in MyD88 gene and protein expression in cardiomyocytes,respectively, whereas Lyz-MyD88−/− had 73 and 67% decrease, respectively, in macrophages (n=3 per group). After lipopolysaccharide treatment, the two groups of MyD88fl/fl littermates had 46% (n=10) and 60% (n=15) of mortality, respectively.Both α-MHC-MyD88−/− and Lyz-MyD88−/− mice had markedly improved survival. Compared with the MyD88fl/fl littermates, Lyz-MyD88−/− mice had warmer body temperature, attenuated systemic and cardiac inflammatory cytokine production,and significantly improved cardiac function, whereas α-MHC-MyD88−/− mice had decreased myocardial inducible nitricoxide synthase induction and modestly preserved cardiac function. CONCLUSIONS: Both cardiomyocyte- and myeloid-MyD88 signaling play a role in cardiac dysfunction and mortality during endotoxin shock. Myeloid-MyD88 signaling plays a predominant role in systemic and cardiac inflammation after endotoxin challenge.


Asunto(s)
Corazón/fisiopatología , Factor 88 de Diferenciación Mieloide/genética , Choque Séptico/genética , Choque Séptico/fisiopatología , Células Madre , Animales , Temperatura Corporal/genética , Eliminación de Gen , Macrófagos/patología , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/patología , Choque Séptico/diagnóstico por imagen , Transducción de Señal/fisiología , Ultrasonografía
17.
J Biol Chem ; 288(41): 29746-59, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-23986437

RESUMEN

The promyelocytic leukemia protein is a well known tumor suppressor, but its role in metabolism is largely unknown. Mice with a deletion in the gene for PML (KO mice) exhibit altered gene expression in liver, adipose tissue, and skeletal muscle, an accelerated rate of fatty acid metabolism, abnormal glucose metabolism, constitutive AMP-activating kinase (AMPK) activation, and insulin resistance in skeletal muscle. Last, an increased rate of energy expenditure protects PML KO mice from the effects of obesity induced by a Western diet. Collectively, our study uncovers a previously unappreciated role of PML in the regulation of metabolism and energy balance in mice.


Asunto(s)
Metabolismo Energético/genética , Proteínas Nucleares/genética , Obesidad/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipoquinas/genética , Tejido Adiposo/metabolismo , Animales , Western Blotting , Temperatura Corporal/genética , Antígenos CD36/genética , Dieta/efectos adversos , Ácidos Grasos/metabolismo , Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Hígado/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Músculo Esquelético/metabolismo , Proteínas Nucleares/deficiencia , Obesidad/etiología , Obesidad/metabolismo , Oxidación-Reducción , Proteína de la Leucemia Promielocítica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/deficiencia , Proteínas Supresoras de Tumor/deficiencia
18.
Eur J Pharmacol ; 719(1-3): 137-144, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-23891845

RESUMEN

The neural substrate of adaptive thermoregulation in mice lacking both brain-type creatine kinase isoforms is further investigated. The cytosolic brain-type creatine kinase (CK-B) and mitochondrial ubiquitous creatine kinase (UbCKmit) are expressed in neural cells throughout the central and peripheral nervous system, where they have an important role in cellular energy homeostasis. Several integral functions appear altered when creatine kinases are absent in the brain (Jost et al., 2002; Streijger et al., 2004, 2005), which has been explained by inefficient neuronal transmission. The CK--/-- double knockout mice demonstrate every morning a body temperature drop of ~1.0 °C, and they have impaired thermogenesis, as revealed by severe hypothermia upon cold exposure. This defective thermoregulation is not associated with abnormal food intake, decreased locomotive activity, or increased torpor sensitivity. Although white and brown adipose tissue fat pads are diminished in CK--/-- mice, intravenous norepinephrine infusion results in a normal brown adipose tissue response with increasing core body temperatures, indicating that the sympathetic innervation functions correctly (Streijger et al., 2009). This study revealed c-fos changes following a cold challenge, and that neuropeptide Y levels were decreased in the paraventricular nucleus of wildtype, but not CK--/--, mice. A reduction in hypothalamic neuropeptide Y is coupled to increased uncoupling protein 1 expression in brown adipose tissue, resulting in thermogenesis. In CK--/-- mice the neuropeptide Y levels did not change. This lack of hypothalamic plasticity of neuropeptide Y might be the result of inefficient neuronal transmission or can be explained by the previous observation of reduced circulating levels of leptin in CK--/-- mice.


Asunto(s)
Regulación de la Temperatura Corporal/genética , Forma BB de la Creatina-Quinasa/deficiencia , Forma Mitocondrial de la Creatina-Quinasa/deficiencia , Técnicas de Inactivación de Genes , Hipotálamo/fisiología , Plasticidad Neuronal/genética , Neuropéptido Y/metabolismo , Animales , Temperatura Corporal/genética , Núcleo Celular/metabolismo , Frío , Forma BB de la Creatina-Quinasa/genética , Forma Mitocondrial de la Creatina-Quinasa/genética , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Ratones , Proteínas Proto-Oncogénicas c-fos/metabolismo
19.
J Anim Sci ; 91(8): 3557-63, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23739793

RESUMEN

Locomotor activity, body temperature, feed intake, and BW were measured on 382 mature male mice sampled from lines previously selected (25 generations) for either high (MH) or low (ML) heat loss and an unselected control (MC). Animals were from all 3 independent replicates of the 3 lines and across 4 generations (68 through 71). Locomotor activity and body temperatures were obtained using implanted transmitters with data collection over 4 d following a 3-d postsurgery recovery period. Data were collected every minute and then averaged into 30-min periods, thus providing 192 data points for each mouse. Least-squares means for feed intake adjusted for BW (Feed/BW, feed·BW(-1)·d(-1), g/g) were 0.1586, 0.1234, and 0.1125 (±0.0022) for MH, MC, and ML, respectively, with line being a highly significant source of variation (P < 0.0003). Line effects for locomotor activity counts, transformed to the 0.25 power for analysis, were significantly different, with MH mice being 2.1 times more active than ML mice (P < 0.003); MC mice were intermediate. Differences in body temperature were significant for both line (P < 0.03) and day effects (P < 0.001), with a 0.32°C difference between the MH and ML lines. Fourier series analysis used the combined significant periodicities of 24, 18, 12, 9, 6, and 3 h to describe circadian cycles for activity and body temperature. All 3 lines expressed daily peaks in body temperature and locomotor activity ∼3 h into darkness and ∼2 h after lights were turned on. There was a stronger relationship between locomotor activity and Feed/BW (P < 0.0001) than between body temperature and Feed/BW (P < 0.01); differences between lines in locomotor activity and body temperature explained 17% and 3%, respectively, of differences between lines in Feed/BW. Thus, line differences in locomotor activity contribute to line differences in maintenance, but approximately 80% of the differences between the MH and ML selection lines in Feed/BW remains independent of differences in locomotor activity.


Asunto(s)
Temperatura Corporal/genética , Ingestión de Alimentos/genética , Ratones/genética , Ratones/fisiología , Actividad Motora/genética , Selección Genética , Animales , Temperatura Corporal/fisiología , Cruzamiento , Ritmo Circadiano , Ingestión de Alimentos/fisiología , Masculino , Actividad Motora/fisiología
20.
J Nutr Biochem ; 24(1): 240-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22901686

RESUMEN

The association of T helper (Th) 1 cells with obesity is well documented in both animals and humans. The T-box transcription factor (T-bet) is known as the transcription factor that is responsible for the development of Th1 cells. However, the role of T-bet in obesity has never been elucidated. The present study aimed to investigate the regulatory function of T-bet on obesity in mice. Th1 cytokine levels were decreased, whereas Th2 cytokine level and GATA-3 messenger RNA (mRNA) expression were increased in T-bet knockout (KO) mice. T-bet KO male mice induced obesity as a result of increased body weight and food efficiency despite the fact that they feed a control diet. T-bet KO mice have an increase in weight of white adipose tissue and levels of triacylglyceride and low-density lipoprotein cholesterol. Interestingly, the expression levels of energy expenditure-related genes were decreased in T-bet KO mice. Both T-bet KO male and female mice had impaired glucose tolerance. In white adipose tissue, leptin, the increase in peroxisome proliferator receptor-γ and CAAT/enhancer-binding protein α mRNA expressions in T-bet KO mice was more than that in wild-type mice. Furthermore, we found that the level of interleukin (IL)-6 mRNA expression in white adipose tissue was elevated in T-bet KO mice but not IL-1ß and tumor necrosis factor-α. IL-6 mRNA expression was increased in adipocyte fraction and stromal vascular fraction in white adipose tissue of T-bet KO mice. Taken together, our results reveal that T-bet may affect obesity through the regulation of IL-6 expression in adipocytes of white adipose tissue.


Asunto(s)
Obesidad/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Adipocitos/fisiología , Tejido Adiposo Blanco/metabolismo , Animales , Temperatura Corporal/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Citocinas/metabolismo , Dieta Alta en Grasa , Femenino , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Interleucina-6/genética , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Noqueados , Obesidad/genética , PPAR gamma/genética , Células TH1/metabolismo , Células Th2/metabolismo , Factor de Necrosis Tumoral alfa/genética , Aumento de Peso/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA