Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.567
Filter
1.
Nat Commun ; 15(1): 3473, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724563

ABSTRACT

Neuronal differentiation-the development of neurons from neural stem cells-involves neurite outgrowth and is a key process during the development and regeneration of neural functions. In addition to various chemical signaling mechanisms, it has been suggested that thermal stimuli induce neuronal differentiation. However, the function of physiological subcellular thermogenesis during neuronal differentiation remains unknown. Here we create methods to manipulate and observe local intracellular temperature, and investigate the effects of noninvasive temperature changes on neuronal differentiation using neuron-like PC12 cells. Using quantitative heating with an infrared laser, we find an increase in local temperature (especially in the nucleus) facilitates neurite outgrowth. Intracellular thermometry reveals that neuronal differentiation is accompanied by intracellular thermogenesis associated with transcription and translation. Suppression of intracellular temperature increase during neuronal differentiation inhibits neurite outgrowth. Furthermore, spontaneous intracellular temperature elevation is involved in neurite outgrowth of primary mouse cortical neurons. These results offer a model for understanding neuronal differentiation induced by intracellular thermal signaling.


Subject(s)
Cell Differentiation , Neurons , Signal Transduction , Temperature , Animals , PC12 Cells , Neurons/physiology , Neurons/cytology , Mice , Rats , Neuronal Outgrowth , Neurogenesis/physiology , Neurites/metabolism , Neurites/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Thermometry/methods , Thermogenesis/physiology
2.
Mol Metab ; 84: 101951, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729241

ABSTRACT

OBJECTIVE: Hypothalamic signals potently stimulate energy expenditure by engaging peripheral mechanisms to restore energy homeostasis. Previous studies have identified several critical hypothalamic sites (e.g. preoptic area (POA) and ventromedial hypothalamic nucleus (VMN)) that could be part of an interconnected neurocircuit that controls tissue thermogenesis and essential for body weight control. However, the key neurocircuit that can stimulate energy expenditure has not yet been established. METHODS: Here, we investigated the downstream mechanisms by which VMN neurons stimulate adipose tissue thermogenesis. We manipulated subsets of VMN neurons acutely as well as chronically and studied its effect on tissue thermogenesis and body weight control, using Sf1Cre and Adcyap1Cre mice and measured physiological parameters under both high-fat diet and standard chow diet conditions. To determine the node efferent to these VMN neurons, that is involved in modulating energy expenditure, we employed electrophysiology and optogenetics experiments combined with measurements using tissue-implantable temperature microchips. RESULTS: Activation of the VMN neurons that express the steroidogenic factor 1 (Sf1; VMNSf1 neurons) reduced body weight, adiposity and increased energy expenditure in diet-induced obese mice. This function is likely mediated, at least in part, by the release of the pituitary adenylate cyclase-activating polypeptide (PACAP; encoded by the Adcyap1 gene) by the VMN neurons, since we previously demonstrated that PACAP, at the VMN, plays a key role in energy expenditure control. Thus, we then shifted focus to the subpopulation of VMNSf1 neurons that contain the neuropeptide PACAP (VMNPACAP neurons). Since the VMN neurons do not directly project to the peripheral tissues, we traced the location of the VMNPACAP neurons' efferents. We identified that VMNPACAP neurons project to and activate neurons in the caudal regions of the POA whereby these projections stimulate tissue thermogenesis in brown and beige adipose tissue. We demonstrated that selective activation of caudal POA projections from VMNPACAP neurons induces tissue thermogenesis, most potently in negative energy balance and activating these projections lead to some similar, but mostly unique, patterns of gene expression in brown and beige tissue. Finally, we demonstrated that the activation of the VMNPACAP neurons' efferents that lie at the caudal POA are necessary for inducing tissue thermogenesis in brown and beige adipose tissue. CONCLUSIONS: These data indicate that VMNPACAP connections with the caudal POA neurons impact adipose tissue function and are important for induction of tissue thermogenesis. Our data suggests that the VMNPACAP → caudal POA neurocircuit and its components are critical for controlling energy balance by activating energy expenditure and body weight control.


Subject(s)
Energy Metabolism , Neurons , Preoptic Area , Thermogenesis , Ventromedial Hypothalamic Nucleus , Animals , Ventromedial Hypothalamic Nucleus/metabolism , Thermogenesis/physiology , Preoptic Area/metabolism , Mice , Neurons/metabolism , Male , Steroidogenic Factor 1/metabolism , Steroidogenic Factor 1/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Diet, High-Fat , Mice, Inbred C57BL , Body Weight , Adipose Tissue, Brown/metabolism
3.
Nat Commun ; 15(1): 2856, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38565851

ABSTRACT

Aging, chronic high-fat diet feeding, or housing at thermoneutrality induces brown adipose tissue (BAT) involution, a process characterized by reduction of BAT mass and function with increased lipid droplet size. Single nuclei RNA sequencing of aged mice identifies a specific brown adipocyte population of Ucp1-low cells that are pyroptotic and display a reduction in the longevity gene syntaxin 4 (Stx4a). Similar to aged brown adipocytes, Ucp1-STX4KO mice display loss of brown adipose tissue mass and thermogenic dysfunction concomitant with increased pyroptosis. Restoration of STX4 expression or suppression of pyroptosis activation protects against the decline in both mass and thermogenic activity in the aged and Ucp1-STX4KO mice. Mechanistically, STX4 deficiency reduces oxidative phosphorylation, glucose uptake, and glycolysis leading to reduced ATP levels, a known triggering signal for pyroptosis. Together, these data demonstrate an understanding of rapid brown adipocyte involution and that physiologic aging and thermogenic dysfunction result from pyroptotic signaling activation.


Subject(s)
Adipose Tissue, Brown , Pyroptosis , Animals , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Signal Transduction , Thermogenesis/physiology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
4.
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
5.
Obesity (Silver Spring) ; 32(6): 1169-1178, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38664956

ABSTRACT

OBJECTIVE: The objective of this study was to compare the magnitude of adaptive thermogenesis (AT), at the level of resting energy expenditure (REE), after a very low-energy diet alone or combined with Roux-en-Y gastric bypass or sleeve gastrectomy, as well as to investigate the association between AT and changes in appetite. METHODS: A total of 44 participants with severe obesity underwent 10 weeks of a very low-energy diet alone or combined with Roux-en-Y gastric bypass or sleeve gastrectomy. Body weight and composition, REE, subjective appetite feelings, and plasma concentrations of gastrointestinal hormones were measured at baseline and week 11. AT, at the level of REE, was defined as a significantly lower measured versus predicted (using a regression model with baseline data) REE. RESULTS: Participants lost 18.4 ± 3.9 kg of body weight and experienced AT, at the level of REE (-121 ± 188 kcal/day; p < 0.001), with no differences among groups. The larger the AT, at the level of REE, the greater the reduction in fasting ghrelin concentrations and the smaller the reduction in feelings of hunger and desire to eat in the postprandial state. CONCLUSIONS: Weight-loss modality does not seem to modulate the magnitude of AT, at the level of REE. The greater the AT, at the level of REE, the greater the drive to eat following weight loss.


Subject(s)
Energy Metabolism , Gastrectomy , Gastric Bypass , Ghrelin , Obesity, Morbid , Thermogenesis , Weight Loss , Humans , Female , Male , Thermogenesis/physiology , Adult , Weight Loss/physiology , Obesity, Morbid/surgery , Obesity, Morbid/diet therapy , Obesity, Morbid/blood , Obesity, Morbid/psychology , Energy Metabolism/physiology , Middle Aged , Ghrelin/blood , Gastrectomy/methods , Appetite/physiology , Diet, Reducing , Adaptation, Physiological , Bariatric Surgery , Basal Metabolism/physiology , Caloric Restriction/methods , Postprandial Period/physiology , Body Composition
6.
Am J Physiol Endocrinol Metab ; 326(5): E696-E708, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38568151

ABSTRACT

Glycogen is a form of energy storage for glucose in different tissues such as liver and skeletal muscle. It remains incompletely understood how glycogen impacts on adipose tissue functionality. Cold exposure elevated the expression of Gys1 that encodes glycogen synthase 1 in brown adipose tissue (BAT) and inguinal white adipose tissue (iWAT). The in vivo function of Gys1 was analyzed using a mouse model in which Gys1 was deleted specifically in adipose tissues. Under normal chow conditions, Gys1 deletion caused little changes to body weight and glucose metabolism. Deletion of Gys1 abrogated upregulation of UCP1 and other thermogenesis-related genes in iWAT upon prolonged cold exposure or treatment with ß3-adrenergic receptor agonist CL-316,243. Stimulation of UCP1 by CL-316,243 in adipose-derived stromal cells (stromal vascular fractions, SVFs) was also reduced by Gys1 deletion. Both the basal glycogen content and CL-316,243-stimulated glycogen accumulation in adipose tissues were reduced by Gys1 deletion. High-fat diet-induced obesity and insulin resistance were aggravated in Gys1-deleted mice. The loss of body weight upon CL-316,243 treatment was also abrogated by the loss of Gys1. In conclusion, our results underscore the pivotal role of glycogen synthesis in adaptive thermogenesis in beige adipose tissue and its impact on diet-induced obesity in mice.NEW & NOTEWORTHY Glycogen is one of major types of fuel reserve in the body and its classical function is to maintain blood glucose level. This study uncovers that glycogen synthesis is required for beige fat tissue to generate heat upon cold exposure. Such a function of glycogen is linked to development of high-fat diet-induced obesity, thus extending our understanding about the physiological functions of glycogen.


Subject(s)
Adipose Tissue, Beige , Diet, High-Fat , Glycogen , Obesity , Thermogenesis , Animals , Thermogenesis/genetics , Thermogenesis/physiology , Mice , Obesity/metabolism , Obesity/genetics , Adipose Tissue, Beige/metabolism , Glycogen/metabolism , Glycogen/biosynthesis , Male , Mice, Knockout , Mice, Inbred C57BL , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Glycogen Synthase/metabolism , Glycogen Synthase/genetics , Cold Temperature , Adaptation, Physiological , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics
7.
Adv Sci (Weinh) ; 11(21): e2306871, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38569495

ABSTRACT

RNA splicing dysregulation and the involvement of specific splicing factors are emerging as common factors in both obesity and metabolic disorders. The study provides compelling evidence that the absence of the splicing factor SRSF1 in mature adipocytes results in whitening of brown adipocyte tissue (BAT) and impaired thermogenesis, along with the inhibition of white adipose tissue browning in mice. Combining single-nucleus RNA sequencing with transmission electron microscopy, it is observed that the transformation of BAT cell types is associated with dysfunctional mitochondria, and SRSF1 deficiency leads to degenerated and fragmented mitochondria within BAT. The results demonstrate that SRSF1 effectively binds to constitutive exon 6 of Ndufs3 pre-mRNA and promotes its inclusion. Conversely, the deficiency of SRSF1 results in impaired splicing of Ndufs3, leading to reduced levels of functional proteins that are essential for mitochondrial complex I assembly and activity. Consequently, this deficiency disrupts mitochondrial integrity, ultimately compromising the thermogenic capacity of BAT. These findings illuminate a novel role for SRSF1 in influencing mitochondrial function and BAT thermogenesis through its regulation of Ndufs3 splicing within BAT.


Subject(s)
Adipocytes, Brown , Homeostasis , Mitochondria , Serine-Arginine Splicing Factors , Thermogenesis , Animals , Thermogenesis/genetics , Thermogenesis/physiology , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Mice , Adipocytes, Brown/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Homeostasis/genetics , Homeostasis/physiology , RNA Splicing/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Male
8.
Pflugers Arch ; 476(5): 769-778, 2024 May.
Article in English | MEDLINE | ID: mdl-38433124

ABSTRACT

Studies have reported enhanced thermoregulatory function as pregnancy progresses; however, it is unclear if differences in thermoregulation are attributed to weight gain or other physiological changes. This study aimed to determine if total body weight will influence thermoregulation (heat production (Hprod)), heart rate, and perceptual measurements in response to weight-bearing exercise during early to late pregnancy. A cross-sectional design of healthy pregnant women at different pregnancy time points (early, T1; middle, T2; late, T3) performed a 7-stage weight-bearing incremental exercise protocol. Measurements of Hprod, HR, and RPE were examined. Two experimental groups were studied: (1) weight matched and (2) non-weight matched, in T1, T2, and T3. During exercise, equivalent Hprod at T1 (326 ± 88 kJ), T2 (330 ± 43 kJ), and T3 (352 ± 52 kJ) (p = 0.504); HR (p = 0.830); and RPE (p = 0.195) were observed in the WM group at each time point. In the NWM group, Hprod (from stages 1-6 of the exercise) increased across pregnancy time points, T1 (291 ± 76 kJ) to T2 (347 ± 41 kJ) and T3 (385 ± 47 kJ) (p < 0.001). HR increased from T1 to T3 in the warm-up to stage 6 (p = 0.009). RPE did not change as pregnancy time point progressed (p = 0.309). Total body weight, irrespective of pregnancy time point, modulates Hprod and HR during exercise. Therefore, accounting for total body weight is crucial when comparing thermoregulatory function during exercise across pregnancy.


Subject(s)
Body Weight , Exercise , Female , Humans , Pregnancy , Exercise/physiology , Adult , Body Weight/physiology , Heart Rate/physiology , Body Temperature Regulation/physiology , Thermogenesis/physiology , Cross-Sectional Studies
9.
J Exp Biol ; 227(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38506250

ABSTRACT

During maximal cold challenge (cold-induced V̇O2,max) in hypoxia, highland deer mice (Peromyscus maniculatus) show higher rates of circulatory fatty acid delivery compared with lowland deer mice. Fatty acid delivery also increases with acclimation to cold hypoxia (CH) and probably plays a major role in supporting the high rates of thermogenesis observed in highland deer mice. However, it is unknown which tissues take up these fatty acids and their relative contribution to thermogenesis. The goal of this study was to determine the uptake of circulating fatty acids into 24 different tissues during hypoxic cold-induced V̇O2,max, by using [1-14C]2-bromopalmitic acid. To uncover evolved and environment-induced changes in fatty acid uptake, we compared lab-born and -raised highland and lowland deer mice, acclimated to either thermoneutral (30°C, 21 kPa O2) or CH (5°C, 12 kPa O2) conditions. During hypoxic cold-induced V̇O2,max, CH-acclimated highlanders decreased muscle fatty acid uptake and increased uptake into brown adipose tissue (BAT) relative to thermoneutral highlanders, a response that was absent in lowlanders. CH acclimation was also associated with increased activities of enzymes citrate synthase and ß-hydroxyacyl-CoA dehydrogenase in the BAT of highlanders, and higher levels of fatty acid translocase CD36 (FAT/CD36) in both populations. This is the first study to show that cold-induced fatty acid uptake is distributed across a wide range of tissues. Highland deer mice show plasticity in this fatty acid distribution in response to chronic cold hypoxia, and combined with higher rates of tissue delivery, this contributes to their survival in the cold high alpine environment.


Subject(s)
Adipose Tissue, Brown , Peromyscus , Animals , Peromyscus/physiology , Fatty Acids , Hypoxia , Acclimatization , Muscles , Thermogenesis/physiology , Cold Temperature
10.
J Sports Sci ; 42(4): 313-322, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38478743

ABSTRACT

In contrast to male football players, research on the nutritional requirements of female football players is limited. This study aimed to assess total daily energy expenditure (TDEE) in professional female football players, along with body composition, physical activity and dietary intake. This observational study included 15 professional football players playing in the highest Dutch Women's League. TDEE was assessed by doubly labelled water over 14 days, along with resting metabolic rate (RMR; ventilated hood), fat-free mass (FFM; dual-energy x-ray absorptiometry), and dietary intake (24-h recalls). Physical activity energy expenditure (PAEE) was derived from subtracting RMR and estimated diet-induced thermogenesis (10%) from TDEE. TDEE was 2882 ± 278 kcal/day (58 ± 5 kcal/kg FFM) and significantly (p < 0.05) correlated with FFM (r = 0.62). PAEE was 1207 ± 213 kcal/d. Weighted energy intake was 2344 kcal [2023-2589]. Carbohydrate intakes were 3.2 ± 0.7, 4.4 ± 1.1 and 5.3 ± 1.9 g/kg body mass for rest, training and match days, respectively, while weighted mean protein intake was 1.9 ± 0.4 g/kg body mass. In conclusion, the energy requirements of professional female football players are moderate to high and can be explained by the substantial PAEE. To fuel these requirements, sports nutritionists should consider shifting the players' focus towards prioritizing adequate carbohydrate intakes, rather than emphasizing high protein consumption.


Subject(s)
Basal Metabolism , Body Composition , Dietary Proteins , Energy Intake , Energy Metabolism , Soccer , Humans , Female , Energy Metabolism/physiology , Energy Intake/physiology , Soccer/physiology , Young Adult , Adult , Dietary Proteins/administration & dosage , Basal Metabolism/physiology , Netherlands , Dietary Carbohydrates/administration & dosage , Nutritional Requirements , Sports Nutritional Physiological Phenomena , Exercise/physiology , Thermogenesis/physiology , Diet
11.
Physiology (Bethesda) ; 39(4): 0, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38536114

ABSTRACT

Whether it is the dramatic suffocating sensation from a heat wave in the summer or the positive reinforcement arising from a hot drink on a cold day; we can certainly agree that our thermal environment underlies our daily rhythms of sensation. Extensive research has focused on deciphering the central circuits responsible for conveying the impact of thermogenesis on mammalian behavior. Here, we revise the recent literature responsible for defining the behavioral correlates that arise from thermogenic fluctuations in mammals. We transition from the physiological significance of thermosensation to the circuitry responsible for the autonomic or behavioral responses associated with it. Subsequently, we delve into the positive and negative valence encoded by thermoregulatory processes. Importantly, we emphasize the crucial junctures where reward, pain, and thermoregulation intersect, unveiling a complex interplay within these neural circuits. Finally, we briefly outline fundamental questions that are pending to be addressed in the field. Fully deciphering the thermoregulatory circuitry in mammals will have far-reaching medical implications. For instance, it may lead to the identification of novel targets to overcome thermal pain or allow the maintenance of our core temperature in prolonged surgeries.


Subject(s)
Body Temperature Regulation , Brain , Cues , Thermosensing , Humans , Animals , Thermosensing/physiology , Brain/physiology , Body Temperature Regulation/physiology , Pain/physiopathology , Thermogenesis/physiology
12.
Int J Mol Sci ; 25(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38339044

ABSTRACT

Spexin (SPX) is a novel adipokine that plays an emerging role in metabolic diseases due to its involvement in carbohydrate homeostasis, weight loss, appetite control, and gastrointestinal movement, among others. In obese patients, SPX plasma levels are reduced. Little is known about the relationship between SPX and white adipose tissue (WAT) thermogenesis. Therefore, the aim of the present study was to evaluate the role of SPX in this process. C57BL/6J male mice were treated or not with SPX for ten days. On day 3, mice were randomly divided into two groups: one kept at room temperature and the other kept at cold temperature (4 °C). Caloric intake and body weight were recorded daily. At the end of the protocol, plasma, abdominal (epididymal), subcutaneous (inguinal), and brown AT (EAT, IAT, and BAT, respectively) depots were collected for measurements. We found that SPX treatment reduced Uncoupling protein 1 levels in WAT under both basal and cold conditions. SPX also reduced cox8b and pgc1α mRNA levels and mitochondrial DNA, principally in IAT. SPX did not modulate the number of beige precursors. SPX decreased spx levels in IAT depots and galr2 in WAT depots. No differences were observed in the BAT depots. In conclusion, we showed, for the first time, that SPX treatment in vivo reduced the thermogenic process in subcutaneous and abdominal AT, being more evident under cold stimulation.


Subject(s)
Adipose Tissue, Brown , Cold Temperature , Peptide Hormones , Thermogenesis , Animals , Humans , Male , Mice , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/physiology , Adipose Tissue, White/metabolism , Mice, Inbred C57BL , Thermogenesis/drug effects , Thermogenesis/physiology , Uncoupling Protein 1/metabolism , Peptide Hormones/pharmacology , Peptide Hormones/physiology
13.
J Acupunct Meridian Stud ; 17(1): 1-11, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38409809

ABSTRACT

Background: : Brown adipose tissue (BAT) is a unique thermogenic tissue in mammals mediated by uncoupling protein 1 (UCP1). The energy generated by glucose and triglyceride metabolism is released and transmitted throughout the body as heat. Understanding the factors influencing BAT function is crucial to determine its metabolic significance and effects on overall health. Although studies have shown that electroacupuncture (EA) at specific acupoints (e.g., ST36) can stimulate BAT, its effects at other acupoints are not well understood. Further research is needed to investigate the potential effects of EA at these acupoints and their association with BAT activation. Objectives: : This study aimed to investigate the effects of EA at the GV20 and EX-HN3 acupoints. Specifically, the effects of EA on BAT thermogenesis were analyzed by infrared thermography, western blotting, and real-time polymerase chain reaction (PCR). Methods: : A total of 12 C57BL/6J mice were randomly divided into the EA and control groups. The EA group received EA at GV20 and EX-HN3 for 20 min once daily for 14 days. The control group underwent the same procedure but without EA. The core body temperature was monitored. Infrared thermal images of the back of each mouse in both groups were captured. BAT samples were collected after euthanasia to analyze UCP1 protein and UCP1 mRNA. Results: : The average skin temperature in the scapular region of the EA group was increased by 1.1℃ compared with that of the C group (p < 0.05). Additionally, the average temperature along the governor vessel in the EA group was increased by 1.6℃ (p = 0.045). EA significantly increased the expression of UCP1 protein (p = 0.001) and UCP1 mRNA (p = 0.002) in BAT, suggesting a potential link between EA and BAT thermogenesis. Conclusion: : EA induced BAT thermogenesis, suggesting GV20 and EX-HN3 as potential acupoints for BAT stimulation. The experimental results also highlighted unique meridian characteristics as demonstrated by elevated skin temperature along the governor vessel in mice.


Subject(s)
Adipose Tissue, Brown , Electroacupuncture , Mice , Animals , Adipose Tissue, Brown/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Mice, Inbred C57BL , Thermogenesis/physiology , RNA, Messenger/metabolism , Mammals/metabolism
14.
Diabetes ; 73(3): 338-347, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38377445

ABSTRACT

The recognition of sensory signals from within the body (interoceptive) and from the external environment (exteroceptive), along with the integration of these cues by the central nervous system, plays a crucial role in maintaining metabolic balance. This orchestration is vital for regulating processes related to both food intake and energy expenditure. Animal model studies indicate that manipulating specific populations of neurons in the central nervous system which influence these processes can effectively modify energy balance. This body of work presents an opportunity for the development of innovative weight loss therapies for the treatment of obesity and type 2 diabetes. In this overview, we delve into the sensory cues and the neuronal populations responsible for their integration, exploring their potential in the development of weight loss treatments for obesity and type 2 diabetes. This article is the first in a series of Perspectives that report on research funded by the American Diabetes Association Pathway to Stop Diabetes program.


Subject(s)
Diabetes Mellitus, Type 2 , Animals , Diabetes Mellitus, Type 2/therapy , Thermogenesis/physiology , Obesity/therapy , Obesity/metabolism , Brain/metabolism , Energy Metabolism/physiology , Weight Loss
15.
Front Med ; 18(2): 205-236, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38165533

ABSTRACT

Macrophages, a heterogeneous population of innate immune cells, exhibit remarkable plasticity and play pivotal roles in coordinating immune responses and maintaining tissue homeostasis within the context of metabolic diseases. The activation of inflammatory macrophages in obese adipose tissue leads to detrimental effects, inducing insulin resistance through increased inflammation, impaired thermogenesis, and adipose tissue fibrosis. Meanwhile, adipose tissue macrophages also play a beneficial role in maintaining adipose tissue homeostasis by regulating angiogenesis, facilitating the clearance of dead adipocytes, and promoting mitochondrial transfer. Exploring the heterogeneity of macrophages in obese adipose tissue is crucial for unraveling the pathogenesis of obesity and holds significant potential for targeted therapeutic interventions. Recently, the dual effects and some potential regulatory mechanisms of macrophages in adipose tissue have been elucidated using single-cell technology. In this review, we present a comprehensive overview of the intricate activation mechanisms and diverse functions of macrophages in adipose tissue during obesity, as well as explore the potential of drug delivery systems targeting macrophages, aiming to enhance the understanding of current regulatory mechanisms that may be potentially targeted for treating obesity or metabolic diseases.


Subject(s)
Adipose Tissue , Macrophages , Obesity , Humans , Obesity/immunology , Macrophages/immunology , Macrophages/physiology , Adipose Tissue/immunology , Inflammation/immunology , Insulin Resistance , Animals , Thermogenesis/physiology , Adipocytes
16.
Arch Biochem Biophys ; 752: 109886, 2024 02.
Article in English | MEDLINE | ID: mdl-38215960

ABSTRACT

Recent studies have shown that some natural compounds from plants prevent obesity and related disorders, including the loss of skeletal muscle mass and strength. In this study, we investigated the effect of echinacoside (ECH), a caffeic acid glycoside from the phenylpropanoid class, on myogenesis and ATP-dependent thermogenesis in the skeletal muscle and its interaction with the dopaminergic receptors 1 and 5 (DRD1 and DRD5). We applied RT-PCR, immunoblot analysis, a staining method, and an assay kit to determine the effects of ECH on diverse target genes and proteins involved in skeletal muscle myogenesis and ATP-consuming futile processes. Our study demonstrated that ECH enhanced myogenic differentiation, glucose, and fatty acid uptake, as well as lipid catabolism, and induced ATP-dependent thermogenesis in vitro and in vivo. Moreover, ECH upregulated mitochondrial biogenesis proteins, mitochondrial oxidative phosphorylation (OXPHOS) complexes, and intracellular Ca2+ signaling as well as thermogenic proteins. These findings were further elucidated by mechanistic studies which showed that ECH mediates myogenesis via the DRD1/5 in C2C12 muscle cells. In addition, ECH stimulates α1-AR-mediated ATP-dependent thermogenesis via the DRD1/5/cAMP/SLN/SERCA1a pathway in C2C12 muscle cells. To the best of our knowledge, this is the first report that demonstrates the myogenic and thermogenic potential of ECH activity through the dopaminergic receptors. Understanding the novel functions of ECH in terms of its ability to prevent skeletal muscle loss and energy expenditure via ATP-consuming futile processes could help to develop potential alternative strategies to address muscle-related diseases, including combating obesity.


Subject(s)
Muscle, Skeletal , Obesity , Humans , Muscle, Skeletal/metabolism , Obesity/metabolism , Glycosides/pharmacology , Adenosine Triphosphate/metabolism , Muscle Development/physiology , Thermogenesis/physiology
17.
Biochem Biophys Res Commun ; 696: 149493, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38219486

ABSTRACT

Brown fat adipose tissue (BAT) is a therapeutic potential target to improve obesity, diabetes and cold acclimation in mammals. During the long-term cold exposure, the hyperplastic sympathetic network is crucial for BAT the maintain the highly thermogenic status. It has been proved that the sympathetic nervous drives the thermogenic activity of BAT via the release of norepinephrine. However, it is still unclear that how the thermogenic BAT affects the remodeling of the hyperplastic sympathetic network, especially during the long-term cold exposure. Here, we showed that following long-term cold exposure, SCD1-mediated monounsaturated fatty acid biosynthesis pathway was enriched, and the ratios of monounsaturated/saturated fatty acids were significantly up-regulated in BAT. And SCD1-deficiency in BAT decreased the capacity of cold acclimation, and suppressed long-term cold mediated BAT thermogenic activation. Furthermore, by using thermoneutral exposure and sympathetic nerve excision models, we disclosed that SCD1-deficiency in BAT affected the thermogenic activity, depended on sympathetic nerve. In mechanism, SCD1-deficiency resulted in the unbalanced ratio of palmitic acid (PA)/palmitoleic acid (PO), with obviously higher level of PA and lower level of PO. And PO supplement efficiently reversed the inhibitory role of SCD1-deficiency on BAT thermogenesis and the hyperplastic sympathetic network. Thus, our data provided insight into the role of SCD1-mediated monounsaturated fatty acids metabolism to the interaction between thermogenic activity BAT and hyperplastic sympathetic networks, and illustrated the critical role of monounsaturated fatty acids biosynthetic pathway in cold acclimation during the long-term cold exposure.


Subject(s)
Adipose Tissue, Brown , Thermogenesis , Animals , Adipose Tissue, Brown/metabolism , Thermogenesis/physiology , Sympathetic Nervous System , Obesity/metabolism , Fatty Acids, Monounsaturated/metabolism , Cold Temperature , Mammals
18.
Immunity ; 57(1): 141-152.e5, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38091996

ABSTRACT

Adipose tissues (ATs) are innervated by sympathetic nerves, which drive reduction of fat mass via lipolysis and thermogenesis. Here, we report a population of immunomodulatory leptin receptor-positive (LepR+) sympathetic perineurial barrier cells (SPCs) present in mice and humans, which uniquely co-express Lepr and interleukin-33 (Il33) and ensheath AT sympathetic axon bundles. Brown ATs (BATs) of mice lacking IL-33 in SPCs (SPCΔIl33) had fewer regulatory T (Treg) cells and eosinophils, resulting in increased BAT inflammation. SPCΔIl33 mice were more susceptible to diet-induced obesity, independently of food intake. Furthermore, SPCΔIl33 mice had impaired adaptive thermogenesis and were unresponsive to leptin-induced rescue of metabolic adaptation. We therefore identify LepR+ SPCs as a source of IL-33, which orchestrate an anti-inflammatory BAT environment, preserving sympathetic-mediated thermogenesis and body weight homeostasis. LepR+IL-33+ SPCs provide a cellular link between leptin and immune regulation of body weight, unifying neuroendocrinology and immunometabolism as previously disconnected fields of obesity research.


Subject(s)
Adipose Tissue, Brown , Leptin , Animals , Humans , Mice , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Body Weight , Energy Metabolism/physiology , Interleukin-33/genetics , Interleukin-33/metabolism , Obesity/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Thermogenesis/physiology
19.
Am J Physiol Regul Integr Comp Physiol ; 326(1): R53-R65, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37955132

ABSTRACT

To maintain heat balance during exercise, humans rely on skin blood flow and sweating to facilitate whole body dry and evaporative heat exchange. These responses are modulated by the rise in body temperature (thermal factors), as well as several nonthermal factors implicated in the cardiovascular response to exercise (i.e., central command, mechanoreceptors, and metaboreceptors). However, the way these nonthermal factors interact with thermal factors to maintain heat balance remains poorly understood. We therefore used direct calorimetry to quantify the effects of dose-dependent increases in the activation of these nonthermal stimuli on whole body dry and evaporative heat exchange during dynamic exercise. In a randomized crossover design, eight participants performed 45-min cycling at a fixed metabolic heat production (200 W/m2) in warm, dry conditions (30°C, 20% relative humidity) on four separate occasions, differing only in the level of lower-limb compression applied via bilateral thigh cuffs pressurized to 0, 30, 60, or 90 mmHg. This model provoked increments in nonthermal activation while ensuring the heat loss required to balance heat production was matched across trials. At end-exercise, dry heat loss was 2 W/m2 [1, 3] lower per 30-mmHg pressure increment (P = 0.006), whereas evaporative heat loss was elevated 5 W/m2 [3, 7] with each pressure increment (P < 0.001). Body heat storage and esophageal temperature did not differ across conditions (both P ≥ 0.600). Our findings indicate that the nonthermal factors engaged during exercise exert dose-dependent, opposing effects on whole body dry and evaporative heat exchange, which do not significantly alter heat balance.NEW & NOTEWORTHY To maintain heat balance during exercise, humans rely on skin blood flow and sweating to facilitate dry and evaporative heat exchange. These responses are modulated by body temperatures (thermal factors) and several nonthermal factors (e.g., central command, metaboreceptors), although the way thermal and nonthermal factors interact to regulate body temperature is poorly understood. We demonstrate that nonthermal factors exert dose-dependent, opposing effects on dry and evaporative heat loss, without altering heat storage during dynamic exercise.


Subject(s)
Body Temperature Regulation , Hot Temperature , Humans , Body Temperature Regulation/physiology , Body Temperature/physiology , Sweating , Thermogenesis/physiology
20.
Obesity (Silver Spring) ; 32(2): 324-338, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37974549

ABSTRACT

OBJECTIVE: Promoting thermogenesis in adipose tissue has been a promising strategy against obesity and related metabolic complications. We aimed to identify compounds that promote thermogenesis in adipocytes and to elucidate their functions and roles in metabolism. METHODS: To identify compounds that directly promote thermogenesis from a structurally diverse set of 4800 compounds, we utilized a cell-based platform for high-throughput screening that induces uncoupling protein 1 (Ucp1) expression in adipocytes. RESULTS: We identified one candidate compound that activates UCP1. Additional characterization of this compound revealed that it induced cellular thermogenesis in adipocytes with negligible cytotoxicity. In a subsequent diet-induced obesity model, mice treated with this compound exhibited a slower rate of weight gain, improved insulin sensitivity, and increased energy expenditure. Mechanistic studies have revealed that this compound increases mitochondrial biogenesis by elevating maximal respiration, which is partly mediated by the protein kinase A (PKA)-p38 mitogen-activated protein kinase (MAPK) signaling pathway. A further comprehensive genetic analysis of adipocytes treated with these compounds identified two novel UCP1-dependent thermogenic genes, potassium voltage-gated channel subfamily C member 2 (Kcnc2) and predicted gene 5627 (Gm5627). CONCLUSIONS: The identified compound can serve as a potential therapeutic drug for the treatment of obesity and its related metabolic disorders. Furthermore, our newly clarified thermogenic genes play an important role in UCP1-dependent thermogenesis in adipocytes.


Subject(s)
Insulin Resistance , Obesity , Uncoupling Protein 1 , Animals , Mice , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Energy Metabolism , Obesity/complications , Obesity/drug therapy , Thermogenesis/physiology , Uncoupling Protein 1/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...