Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 121(19): e2311116121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38683977

ABSTRACT

Conventionally, women are perceived to feel colder than men, but controlled comparisons are sparse. We measured the response of healthy, lean, young women and men to a range of ambient temperatures typical of the daily environment (17 to 31 °C). The Scholander model of thermoregulation defines the lower critical temperature as threshold of the thermoneutral zone, below which additional heat production is required to defend core body temperature. This parameter can be used to characterize the thermoregulatory phenotypes of endotherms on a spectrum from "arctic" to "tropical." We found that women had a cooler lower critical temperature (mean ± SD: 21.9 ± 1.3 °C vs. 22.9 ± 1.2 °C, P = 0.047), resembling an "arctic" shift compared to men. The more arctic profile of women was predominantly driven by higher insulation associated with more body fat compared to men, countering the lower basal metabolic rate associated with their smaller body size, which typically favors a "tropical" shift. We did not detect sex-based differences in secondary measures of thermoregulation including brown adipose tissue glucose uptake, muscle electrical activity, skin temperatures, cold-induced thermogenesis, or self-reported thermal comfort. In conclusion, the principal contributors to individual differences in human thermoregulation are physical attributes, including body size and composition, which may be partly mediated by sex.


Subject(s)
Body Temperature Regulation , Humans , Female , Male , Body Temperature Regulation/physiology , Adult , Arctic Regions , Young Adult , Adipose Tissue, Brown/physiology , Adipose Tissue, Brown/metabolism , Sex Characteristics , Sex Factors , Body Temperature/physiology , Thermogenesis/physiology , Basal Metabolism/physiology
2.
Purinergic Signal ; 19(3): 551-564, 2023 09.
Article in English | MEDLINE | ID: mdl-36781825

ABSTRACT

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.


Subject(s)
Adenosine , Hypothermia , Mice , Animals , Adenosine/pharmacology , Hypothermia/chemically induced , Nimodipine/adverse effects , Receptors, Purinergic P1 , Dipyridamole/adverse effects
3.
PLoS Biol ; 17(3): e3000161, 2019 03.
Article in English | MEDLINE | ID: mdl-30822301

ABSTRACT

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.


Subject(s)
Hypothermia/metabolism , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/metabolism , Receptor, Adenosine A3/metabolism , Animals , Blood Pressure/genetics , Blood Pressure/physiology , Body Temperature/genetics , Body Temperature/physiology , Caffeine/pharmacology , Female , Genotype , Heart Rate/genetics , Heart Rate/physiology , Hypothermia/chemically induced , Hypothermia/genetics , Inosine/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Knockout , Phenotype , Receptor, Adenosine A1/genetics , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A3/genetics , Uridine/toxicity
5.
Am J Physiol Endocrinol Metab ; 319(2): E438-E446, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32691633

ABSTRACT

Understanding mouse thermal physiology informs the usefulness of mice as models of human disease. It is widely assumed that the mouse tail contributes greatly to heat loss (as it does in rat), but this has not been quantitated. We studied C57BL/6J mice after tail amputation. Tailless mice housed at 22°C did not differ from littermate controls in body weight, lean or fat content, or energy expenditure. With acute changes in ambient temperature from 19 to 39°C, tailless and control mice demonstrated similar body temperatures (Tb), metabolic rates, and heat conductances and no difference in thermoneutral point. Treatment with prazosin, an α1-adrenergic antagonist and vasodilator, increased tail temperature in control mice by up to 4.8 ± 0.8°C. Comparing prazosin treatment in tailless and control mice suggested that the tail's contribution to total heat loss was a nonsignificant 3.4%. Major heat stress produced by treatment at 30°C with CL316243, a ß3-adrenergic agonist, increased metabolic rate and Tb and, at a matched increase in metabolic rate, the tailless mice showed a 0.72 ± 0.14°C greater Tb increase and 7.6% lower whole body heat conductance. Thus, the mouse tail is a useful biomarker of vasodilation and thermoregulation, but in our experiments contributes only 5-8% of whole body heat dissipation, less than the 17% reported for rat. Heat dissipation through the tail is important under extreme scenarios such as pharmacological activation of brown adipose tissue; however, non-tail contributions to heat loss may have been underestimated in the mouse.


Subject(s)
Body Temperature Regulation/physiology , Models, Animal , Tail/physiology , Adrenergic alpha-1 Receptor Antagonists , Amputation, Surgical , Animals , Body Composition/physiology , Body Surface Area , Body Temperature Regulation/drug effects , Body Weight/physiology , Energy Metabolism/physiology , Heat-Shock Response , Mice , Mice, Inbred C57BL , Prazosin/pharmacology , Rats , Tail/surgery , Vasodilation/physiology
6.
Am J Physiol Endocrinol Metab ; 315(3): E357-E366, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29812984

ABSTRACT

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.


Subject(s)
Histamine Agonists/pharmacology , Hypothermia/chemically induced , Mast Cells/drug effects , Peptides, Cyclic/pharmacology , alpha-MSH/analogs & derivatives , Animals , Histamine Release/drug effects , Histamine Release/genetics , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , alpha-MSH/pharmacology
7.
Am J Hum Genet ; 92(5): 827-34, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23643386

ABSTRACT

The amount of weight loss attained after Roux-en-Y gastric bypass (RYGB) surgery follows a wide and normal distribution, and recent evidence indicates that this weight loss is due to physiological, rather than mechanical, mechanisms. To identify potential genetic factors associated with weight loss after RYGB, we performed a genome-wide association study (GWAS) of 693 individuals undergoing RYGB and then replicated this analysis in an independent population of 327 individuals undergoing RYGB. We found that a 15q26.1 locus near ST8SIA2 and SLCO3A1 was significantly associated with weight loss after RYGB. Expression of ST8SIA2 in omental fat of these individuals at baseline was significantly associated with weight loss after RYGB. Gene expression analysis in RYGB and weight-matched, sham-operated (WMS) mice revealed that expression of St8sia2 and Slco3a1 was significantly altered in metabolically active tissues in RYGB-treated compared to WMS mice. These findings provide strong evidence for specific genetic influences on weight loss after RYGB and underscore the biological nature of the response to RYGB.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , Gastric Bypass , Sialyltransferases/genetics , Weight Loss/genetics , Animals , Aquaporins/genetics , Genome-Wide Association Study , Humans , Linear Models , Mice , Organic Anion Transporters/genetics , Polymorphism, Single Nucleotide/genetics
8.
Am J Physiol Heart Circ Physiol ; 310(7): H891-8, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26801314

ABSTRACT

Bombesin-like receptor 3 (BRS-3) is an orphan G protein-coupled receptor that regulates energy expenditure, food intake, and body weight. We examined the effects of BRS-3 deletion and activation on blood pressure and heart rate. In free-living, telemetered Brs3 null mice the resting heart rate was 10% lower than wild-type controls, while the resting mean arterial pressure was unchanged. During physical activity, the heart rate and blood pressure increased more in Brs3 null mice, reaching a similar heart rate and higher mean arterial pressure than control mice. When sympathetic input was blocked with propranolol, the heart rate of Brs3 null mice was unchanged, while the heart rate in control mice was reduced to the level of the null mice. The intrinsic heart rate, measured after both sympathetic and parasympathetic blockade, was similar in Brs3 null and control mice. Intravenous infusion of the BRS-3 agonist MK-5046 increased mean arterial pressure and heart rate in wild-type but not in Brs3 null mice, and this increase was blocked by pretreatment with clonidine, a sympatholytic, centrally acting α2-adrenergic agonist. In anesthetized mice, hypothalamic infusion of MK-5046 also increased both mean arterial pressure and heart rate. Taken together, these data demonstrate that BRS-3 contributes to resting cardiac sympathetic tone, but is not required for activity-induced increases in heart rate and blood pressure. The data suggest that BRS-3 activation increases heart rate and blood pressure via a central sympathetic mechanism.


Subject(s)
Blood Pressure , Heart Rate , Receptors, Bombesin/metabolism , Sympathetic Nervous System/physiology , Adrenergic Agents/pharmacology , Animals , Mice , Mice, Inbred C57BL , Receptors, Bombesin/genetics , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
9.
J Pharmacol Exp Ther ; 356(2): 474-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26606937

ABSTRACT

Adenosine can induce hypothermia, as previously demonstrated for adenosine A1 receptor (A1AR) agonists. Here we use the potent, specific A3AR agonists MRS5698, MRS5841, and MRS5980 to show that adenosine also induces hypothermia via the A3AR. The hypothermic effect of A3AR agonists is independent of A1AR activation, as the effect was fully intact in mice lacking A1AR but abolished in mice lacking A3AR. A3AR agonist-induced hypothermia was attenuated by mast cell granule depletion, demonstrating that the A3AR hypothermia is mediated, at least in part, via mast cells. Central agonist dosing had no clear hypothermic effect, whereas peripheral dosing of a non-brain-penetrant agonist caused hypothermia, suggesting that peripheral A3AR-expressing cells drive the hypothermia. Mast cells release histamine, and blocking central histamine H1 (but not H2 or H4) receptors prevented the hypothermia. The hypothermia was preceded by hypometabolism and mice with hypothermia preferred a cooler environmental temperature, demonstrating that the hypothermic state is a coordinated physiologic response with a reduced body temperature set point. Importantly, hypothermia is not required for the analgesic effects of A3AR agonists, which occur with lower agonist doses. These results support a mechanistic model for hypothermia in which A3AR agonists act on peripheral mast cells, causing histamine release, which stimulates central histamine H1 receptors to induce hypothermia. This mechanism suggests that A3AR agonists will probably not be useful for clinical induction of hypothermia.


Subject(s)
Hypothermia/metabolism , Receptor, Adenosine A3/metabolism , Receptors, Histamine H1/metabolism , Adenosine A3 Receptor Agonists/pharmacology , Animals , Dose-Response Relationship, Drug , Hypothermia/chemically induced , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL
10.
J Biol Chem ; 289(28): 19341-50, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24872412

ABSTRACT

The chemical uncoupler 2,4-dinitrophenol (DNP) was an effective and widely used weight loss drug in the early 1930s. However, the physiology of DNP has not been studied in detail because toxicity, including hyperthermia and death, reduced interest in the clinical use of chemical uncouplers. To investigate DNP action, mice fed a high fat diet and housed at 30 °C (to minimize facultative thermogenesis) were treated with 800 mg/liter DNP in drinking water. DNP treatment increased energy expenditure by ∼ 17%, but did not change food intake. DNP-treated mice weighed 26% less than controls after 2 months of treatment due to decreased fat mass, without a change in lean mass. DNP improved glucose tolerance and reduced hepatic steatosis without observed toxicity. DNP treatment also reduced circulating T3 and T4 levels, Ucp1 expression, and brown adipose tissue activity, demonstrating that DNP-mediated heat generation substituted for brown adipose tissue thermogenesis. At 22 °C, a typical vivarium temperature that is below thermoneutrality, DNP treatment had no effect on body weight, adiposity, or glucose homeostasis. Thus, environmental temperature should be considered when assessing an anti-obesity drug in mice, particularly agents acting on energy expenditure. Furthermore, the beneficial effects of DNP suggest that chemical uncouplers deserve further investigation for the treatment of obesity and its comorbidities.


Subject(s)
2,4-Dinitrophenol/pharmacology , Adiposity/drug effects , Body Temperature Regulation/drug effects , Diet/adverse effects , Energy Metabolism/drug effects , Obesity/drug therapy , Uncoupling Agents/pharmacology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Animals , Eating/drug effects , Gene Expression Regulation/drug effects , Ion Channels/biosynthesis , Mice , Mitochondrial Proteins/biosynthesis , Obesity/chemically induced , Obesity/metabolism , Obesity/pathology , Uncoupling Protein 1
11.
Am J Physiol Endocrinol Metab ; 306(6): E681-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24452453

ABSTRACT

Bombesin receptor subtype-3 (BRS-3) regulates energy homeostasis, with Brs3 knockout (Brs3(-/y)) mice being hypometabolic, hypothermic, and hyperphagic and developing obesity. We now report that the reduced body temperature is more readily detected if body temperature is analyzed as a function of physical activity level and light/dark phase. Physical activity level correlated best with body temperature 4 min later. The Brs3(-/y) metabolic phenotype is not due to intrinsically impaired brown adipose tissue function or in the communication of sympathetic signals from the brain to brown adipose tissue, since Brs3(-/y) mice have intact thermogenic responses to stress, acute cold exposure, and ß3-adrenergic activation, and Brs3(-/y) mice prefer a cooler environment. Treatment with the BRS-3 agonist MK-5046 increased brown adipose tissue temperature and body temperature in wild-type but not Brs3(-/y) mice. Intrahypothalamic infusion of MK-5046 increased body temperature. These data indicate that the BRS-3 regulation of body temperature is via a central mechanism, upstream of sympathetic efferents. The reduced body temperature in Brs3(-/y) mice is due to altered regulation of energy homeostasis affecting higher center regulation of body temperature, rather than an intrinsic defect in brown adipose tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , Body Temperature Regulation , Hypothalamus/metabolism , Neurons/metabolism , Receptors, Bombesin/metabolism , Sympathetic Nervous System/metabolism , Thermogenesis , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/innervation , Adrenergic beta-3 Receptor Agonists/administration & dosage , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Body Temperature Regulation/drug effects , Cold-Shock Response/drug effects , Crosses, Genetic , Dioxoles/administration & dosage , Dioxoles/pharmacology , Efferent Pathways/drug effects , Efferent Pathways/metabolism , Energy Metabolism/drug effects , Hypothalamus/drug effects , Imidazoles/administration & dosage , Imidazoles/pharmacology , Infusions, Intravenous , Infusions, Intraventricular , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Receptors, Bombesin/agonists , Receptors, Bombesin/genetics , Sympathetic Nervous System/drug effects , Thermogenesis/drug effects
14.
Genome Res ; 21(7): 1008-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21602305

ABSTRACT

To map the genetics of gene expression in metabolically relevant tissues and investigate the diversity of expression SNPs (eSNPs) in multiple tissues from the same individual, we collected four tissues from approximately 1000 patients undergoing Roux-en-Y gastric bypass (RYGB) and clinical traits associated with their weight loss and co-morbidities. We then performed high-throughput genotyping and gene expression profiling and carried out a genome-wide association analyses for more than 100,000 gene expression traits representing four metabolically relevant tissues: liver, omental adipose, subcutaneous adipose, and stomach. We successfully identified 24,531 eSNPs corresponding to about 10,000 distinct genes. This represents the greatest number of eSNPs identified to our knowledge by any study to date and the first study to identify eSNPs from stomach tissue. We then demonstrate how these eSNPs provide a high-quality disease map for each tissue in morbidly obese patients to not only inform genetic associations identified in this cohort, but in previously published genome-wide association studies as well. These data can aid in elucidating the key networks associated with morbid obesity, response to RYGB, and disease as a whole.


Subject(s)
Gastric Mucosa/metabolism , Liver/metabolism , Obesity, Morbid/epidemiology , Obesity, Morbid/genetics , Adiposity/genetics , Adult , Cohort Studies , Comorbidity , Databases, Genetic , Female , Gastric Bypass , Gene Expression Profiling , Genome-Wide Association Study/methods , Genotype , Humans , Male , Middle Aged , Obesity, Morbid/surgery , Polymorphism, Single Nucleotide , Weight Loss
15.
Nat Methods ; 9(1): 57-63, 2011 Dec 28.
Article in English | MEDLINE | ID: mdl-22205519

ABSTRACT

We present a consolidated view of the complexity and challenges of designing studies for measurement of energy metabolism in mouse models, including a practical guide to the assessment of energy expenditure, energy intake and body composition and statistical analysis thereof. We hope this guide will facilitate comparisons across studies and minimize spurious interpretations of data. We recommend that division of energy expenditure data by either body weight or lean body weight and that presentation of group effects as histograms should be replaced by plotting individual data and analyzing both group and body-composition effects using analysis of covariance (ANCOVA).


Subject(s)
Energy Intake , Energy Metabolism , Mice/physiology , Animals , Body Composition , Environment , Housing, Animal , Mice, Mutant Strains/genetics , Obesity/etiology , Phenotype
16.
Nature ; 452(7186): 423-8, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18344981

ABSTRACT

Common human diseases result from the interplay of many genes and environmental factors. Therefore, a more integrative biology approach is needed to unravel the complexity and causes of such diseases. To elucidate the complexity of common human diseases such as obesity, we have analysed the expression of 23,720 transcripts in large population-based blood and adipose tissue cohorts comprehensively assessed for various phenotypes, including traits related to clinical obesity. In contrast to the blood expression profiles, we observed a marked correlation between gene expression in adipose tissue and obesity-related traits. Genome-wide linkage and association mapping revealed a highly significant genetic component to gene expression traits, including a strong genetic effect of proximal (cis) signals, with 50% of the cis signals overlapping between the two tissues profiled. Here we demonstrate an extensive transcriptional network constructed from the human adipose data that exhibits significant overlap with similar network modules constructed from mouse adipose data. A core network module in humans and mice was identified that is enriched for genes involved in the inflammatory and immune response and has been found to be causally associated to obesity-related traits.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation/genetics , Obesity/genetics , Adipose Tissue/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Blood/metabolism , Body Mass Index , Cohort Studies , Female , Genome, Human , Humans , Iceland , Lod Score , Male , Mice , Middle Aged , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Sample Size , Waist-Hip Ratio , White People/genetics
17.
Mol Metab ; 84: 101946, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657735

ABSTRACT

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.


Subject(s)
Body Temperature , Circadian Rhythm , Energy Metabolism , Photoperiod , Ultradian Rhythm , Animals , Mice , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/physiology , Body Temperature/physiology , Circadian Rhythm/physiology , Energy Metabolism/physiology , Sleep/physiology , Ultradian Rhythm/physiology , Wakefulness/physiology
18.
Cell Rep ; 43(8): 114501, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39067024

ABSTRACT

Evaluation of weight loss drugs is usually performed in diet-induced obese mice housed at ∼22°C. This is a cold stress that increases energy expenditure by ∼35% compared to thermoneutrality (∼30°C), which may overestimate drug-induced weight loss. We investigated five anti-obesity mechanisms that have been in clinical development, comparing weight loss in mice housed at 22°C vs. 30°C. Glucagon-like peptide-1 (GLP-1), human fibroblast growth factor 21 (hFGF21), and melanocortin-4 receptor (MC4R) agonist induced similar weight losses. Peptide YY elicited greater vehicle-subtracted weight loss at 30°C (7.2% vs. 1.4%), whereas growth differentiation factor 15 (GDF15) was more effective at 22°C (13% vs. 6%). Independent of ambient temperature, GLP-1 and hFGF21 prevented the reduction in metabolic rate caused by weight loss. There was no simple rule for a better prediction of human drug efficacy based on ambient temperature, but since humans live at thermoneutrality, drug testing using mice should include experiments near thermoneutrality.


Subject(s)
Weight Loss , Animals , Humans , Weight Loss/drug effects , Mice , Male , Glucagon-Like Peptide 1/metabolism , Fibroblast Growth Factors/metabolism , Mice, Inbred C57BL , Obesity/metabolism , Housing, Animal , Temperature , Receptor, Melanocortin, Type 4/metabolism , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use
19.
bioRxiv ; 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39091749

ABSTRACT

Across mammalian species, new mothers undergo considerable behavioral changes to nurture their offspring and meet the caloric demands of milk production1-5. While many neural circuits underlying feeding and parenting behaviors are well characterized6-9, it is unclear how these different circuits interact and adapt during lactation. Here, we characterized the transcriptomic changes in the arcuate nucleus (ARC) and the medial preoptic area (MPOA) of the mouse hypothalamus in response to lactation and hunger. Furthermore, we showed that heightened appetite in lactating mice was accompanied by increased activity of hunger-promoting agouti-related peptide (AgRP) neurons in the ARC. To assess the strength of hunger versus maternal drives, we designed a conflict assay where female mice chose between a food source or a chamber containing pups and nesting material. Although food-deprived lactating mothers prioritized parenting over feeding, hunger reduced the duration and disrupted the sequences of parenting behaviors in both lactating and virgin females. We discovered that ARCAgRP neurons directly inhibit bombesin receptor subtype-3 (BRS3) neurons in the MPOA, a population that governs both parenting and satiety. Selective activation of this ARCAgRP to MPOABRS3 circuit shifted behaviors from parenting to food-seeking. Thus, hypothalamic networks are modulated by physiological states and work antagonistically during the prioritization of competing motivated behaviors.

20.
J Pharmacol Exp Ther ; 347(1): 100-16, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23892571

ABSTRACT

Bombesin-receptor-subtype-3 (BRS-3) is an orphan G-protein-coupled receptor of the bombesin (Bn) family whose natural ligand is unknown and which does not bind any natural Bn-peptide with high affinity. It is present in the central nervous system, peripheral tissues, and tumors; however, its role in normal physiology/pathophysiology is largely unknown because of the lack of selective ligands. Recently, MK-5046 [(2S)-1,1,1-trifluoro-2-[4-(1H-pyrazol-1-yl)phenyl]-3-(4-{[1-(trifluoromethyl)cyclopropyl]methyl}-1H-imidazol-2-yl)propan-2-ol] and Bantag-1 [Boc-Phe-His-4-amino-5-cyclohexyl-2,4,5-trideoxypentonyl-Leu-(3-dimethylamino) benzylamide N-methylammonium trifluoroacetate], a nonpeptide agonist and a peptide antagonist, respectively, for BRS-3 have been described, but there have been limited studies on their pharmacology. We studied MK-5046 and Bantag-1 interactions with human Bn-receptors-human bombesin receptor subtype-3 (hBRS-3), gastrin-releasing peptide receptor (GRP-R), and neuromedin B receptor (NMB-R)-and compared them with the nonselective, peptide-agonist [d-Tyr6,ßAla11,Phe13,Nle14]Bn-(6-14) (peptide #1). Receptor activation was detected by activation of phospholipase C (PLC), mitogen-activated protein kinase (MAPK), focal adhesion kinase (FAK), paxillin, and Akt. In hBRS-3 cells, the relative affinities were Bantag-1 (1.3 nM) > peptide #1 (2 nM) > MK-5046 (37-160 nM) > GRP, NMB (>10 µM), and the binding-dose-inhibition curves were broad (>4 logs), with Hill coefficients differing significantly from unity. Curve-fitting demonstrated high-affinity (MK-5046, Ki = 0.08 nM) and low-affinity (MK-5046, Ki = 11-29 nM) binding sites. For PLC activation in hBRS-3 cells, the relative potencies were MK-5046 (0.02 nM) > peptide #1 (6 nM) > GRP, NMB, Bantag-1 (>10 µM), and MK-5046 had a biphasic dose response, whereas peptide #1 was monophasic. Bantag-1 was a specific hBRS-3-antagonist. In hBRS-3 cells, MK-5046 was a full agonist for activation of MAPK, FAK, Akt, and paxillin; however, it was a partial agonist for phospholipase A2 (PLA2) activation. The kinetics of activation/duration of action for PLC/MAPK activation of MK-5046 and peptide #1 differed, with peptide #1 causing more rapid stimulation; however, MK-5046 had more prolonged activity. Our study finds that MK-5046 and Bantag-1 have high affinity/selectivity for hBRS-3. The nonpeptide MK-5046 and peptide #1 agonists differ markedly in their receptor coupling, ability to activate different signaling cascades, and kinetics/duration of action. These results show that their hBRS-3 receptor activation is not always concordant and could lead to markedly different cellular responses.


Subject(s)
Imidazoles/pharmacology , Pyrazoles/pharmacology , Receptors, Bombesin/agonists , Receptors, Bombesin/antagonists & inhibitors , 3T3 Cells , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Imidazoles/metabolism , Mice , Mice, Inbred BALB C , Oligopeptides/chemistry , Oligopeptides/metabolism , Oligopeptides/pharmacology , Pyrazoles/metabolism , Receptors, Bombesin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL