Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.065
Filter
1.
Commun Biol ; 7(1): 793, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951146

ABSTRACT

Brown and brown-like adipose tissues have attracted significant attention for their role in metabolism and therapeutic potential in diabetes and obesity. Despite compelling evidence of an interplay between adipocytes and lymphocytes, the involvement of these tissues in immune responses remains largely unexplored. This study explicates a newfound connection between neuroinflammation and brown- and bone marrow adipose tissue. Leveraging the use of [18F]F-AraG, a mitochondrial metabolic tracer capable of tracking activated lymphocytes and adipocytes simultaneously, we demonstrate, in models of glioblastoma and multiple sclerosis, the correlation between intracerebral immune infiltration and changes in brown- and bone marrow adipose tissue. Significantly, we show initial evidence that a neuroinflammation-adipose tissue link may also exist in humans. This study proposes the concept of an intricate immuno-neuro-adipose circuit, and highlights brown- and bone marrow adipose tissue as an intermediary in the communication between the immune and nervous systems. Understanding the interconnectedness within this circuitry may lead to advancements in the treatment and management of various conditions, including cancer, neurodegenerative diseases and metabolic disorders.


Subject(s)
Adipose Tissue, Brown , Neuroinflammatory Diseases , Animals , Humans , Adipose Tissue, Brown/metabolism , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Bone Marrow/metabolism , Mice , Male , Glioblastoma/pathology , Glioblastoma/immunology , Glioblastoma/metabolism , Mice, Inbred C57BL , Female , Multiple Sclerosis/pathology , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/diagnostic imaging , Positron-Emission Tomography
2.
J Nutr Sci Vitaminol (Tokyo) ; 70(3): 193-202, 2024.
Article in English | MEDLINE | ID: mdl-38945884

ABSTRACT

Oleuropein aglycone (OA), which is the absorbed form of oleuropein, is a major phenolic compound in extra virgin olive oil. We analyzed the anti-obesity effect of OA intake combined with mild treadmill walking (MTW, 4 m/min for 20 min/d, 5-6 d/wk, without electric shocks and slope) in rats under a high-fat diet (HF). Four-week-old male Sprague-Dawley rats (n=28) were equally divided into four groups: control (HF), 0.08% oleuropein-supplemented HF (HFO), HF with MTW (HF+W), and HFO with MTW (HFO+W) groups. After 28 d, the inguinal subcutaneous fat content and weight gain were significantly lower in the HFO+W group than in the control group. The HFO+W group also had significantly higher levels of urinary noradrenaline secretion, interscapular brown adipose tissue, uncoupling protein 1, brain transient receptor potential ankyrin subtype 1 (TRPA1), vanilloid subtype 1 (TRPV1), and brain-derived neurotrophic factor (BDNF) than the control group. Especially, the HFO+W group showed a synergistic effect on noradrenaline secretion. Therefore, OA combined with MTW may accelerate the enhancement of UCP1 and BDNF levels in rats with HF-induced obesity by increasing noradrenaline secretion after TRPA1 and TRPV1 activation.


Subject(s)
Adipose Tissue, Brown , Brain-Derived Neurotrophic Factor , Diet, High-Fat , Iridoid Glucosides , Iridoids , Norepinephrine , Obesity , Rats, Sprague-Dawley , TRPA1 Cation Channel , Uncoupling Protein 1 , Animals , Male , Uncoupling Protein 1/metabolism , Iridoid Glucosides/pharmacology , Obesity/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Iridoids/pharmacology , Norepinephrine/metabolism , TRPA1 Cation Channel/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Rats , Anti-Obesity Agents/pharmacology , Walking , Weight Gain/drug effects , Physical Conditioning, Animal , TRPV Cation Channels
3.
Biomolecules ; 14(6)2024 May 23.
Article in English | MEDLINE | ID: mdl-38927022

ABSTRACT

Recent studies increasingly suggest that targeting brown/beige adipose tissues to enhance energy expenditure offers a novel therapeutic approach for treating metabolic diseases. Brown/beige adipocytes exhibit elevated expression of uncoupling protein 1 (UCP1), which is a thermogenic protein that efficiently converts energy into heat, particularly in response to cold stimulation. Polyphenols possess potential anti-obesity properties, but their pharmacological effects are limited by their bioavailability and distribution within tissue. This study discovered 18a, a polyphenol compound with a favorable distribution within adipose tissues, which transcriptionally activates UCP1, thereby promoting thermogenesis and enhancing mitochondrial respiration in brown adipocytes. Furthermore, in vivo studies demonstrated that 18a prevents high-fat-diet-induced weight gain and improves insulin sensitivity. Our research provides strong mechanistic evidence that UCP1 is a complex mediator of 18a-induced thermogenesis, which is a critical process in obesity mitigation. Brown adipose thermogenesis is triggered by 18a via the AMPK-PGC-1α pathway. As a result, our research highlights a thermogenic controlled polyphenol compound 18a and clarifies its underlying mechanisms, thus offering a potential strategy for the thermogenic targeting of adipose tissue to reduce the incidence of obesity and its related metabolic problems.


Subject(s)
Diet, High-Fat , Obesity , Polyphenols , Thermogenesis , Uncoupling Protein 1 , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Thermogenesis/drug effects , Animals , Obesity/metabolism , Obesity/drug therapy , Polyphenols/pharmacology , Mice , Diet, High-Fat/adverse effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Male , Mice, Inbred C57BL , Humans , Energy Metabolism/drug effects
4.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38928011

ABSTRACT

Adipose tissue is conventionally recognized as a metabolic organ responsible for storing energy. However, a proportion of adipose tissue also functions as a thermogenic organ, contributing to the inhibition of weight gain and prevention of metabolic diseases. In recent years, there has been significant progress in the study of thermogenic fats, particularly brown adipose tissue (BAT). Despite this progress, the mechanism underlying thermogenesis in beige adipose tissue remains highly controversial. It is widely acknowledged that beige adipose tissue has three additional thermogenic mechanisms in addition to the conventional UCP1-dependent thermogenesis: Ca2+ cycling thermogenesis, creatine substrate cycling thermogenesis, and triacylglycerol/fatty acid cycling thermogenesis. This paper delves into these three mechanisms and reviews the latest advancements in the molecular regulation of thermogenesis from the molecular genetic perspective. The objective of this review is to provide readers with a foundation of knowledge regarding the beige fats and a foundation for future research into the mechanisms of this process, which may lead to the development of new strategies for maintaining human health.


Subject(s)
Adipocytes, Beige , Thermogenesis , Thermogenesis/genetics , Humans , Adipocytes, Beige/metabolism , Animals , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Adipose Tissue, Brown/metabolism , Energy Metabolism , Calcium/metabolism , Fatty Acids/metabolism , Triglycerides/metabolism , Adipose Tissue, Beige/metabolism
5.
STAR Protoc ; 5(2): 103042, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38850537

ABSTRACT

The mitochondrial stress test is a gold-standard approach for assessing adipose tissue physiological functions and pathological changes. Here, we present a protocol for conducting Seahorse assays using ex vivo mouse brown and white adipose depots. We describe steps for rehydrating the cartridge, preparing freshly harvested fat depots, placing them onto an islet capture plate, and incubating them in a non-CO2 incubator. We then detail procedures for adding mitochondrial stressor solutions and conducting the mitochondrial stress test using the Seahorse XFe24 Analyzer. For complete details on the use and execution of this protocol, please refer to An et al.1.


Subject(s)
Adipose Tissue, Brown , Adipose Tissue, White , Mitochondria , Animals , Mice , Adipose Tissue, White/metabolism , Adipose Tissue, Brown/metabolism , Mitochondria/metabolism
6.
J Endocrinol ; 262(2)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38842921

ABSTRACT

Characteristic symptoms of hyperthyroidism include weight loss, heart palpitation, and sweating. Thyroid hormones (TH) can stimulate thermogenesis through central and peripheral mechanisms. Previous studies have shown an association between dysfunction of cardiotrophin-like cytokine factor 1 (CLCF1) and cold-induced sweating syndrome, with recent research also indicating a link between CLCF1 and brown adipose tissue thermogenesis. However, it remains unclear whether CLCF1 and TH have synergistic or antagonistic effects on thermogenesis. This study aims to investigate the influence of thyroid hormone on circulating CLCF1 levels in humans and explore the potential possibilities of thyroid hormone in regulating energy metabolism by modulating Clcf1 in mice. By recruiting hyperthyroid patients and healthy subjects, we observed significantly lower serum CLCF1 levels in hyperthyroid patients compared to healthy subjects, with serum CLCF1 levels independently associated with hyperthyroidism after adjusting for potential confounders. Tissue analysis from mice treated with T3 revealed a decrease in CLCF1 expression in BAT and iWAT of C57BL/6 mice. These findings suggest that TH may play a role in regulating CLCF1 expression in adipose tissue.


Subject(s)
Hyperthyroidism , Mice, Inbred C57BL , Triiodothyronine , Hyperthyroidism/blood , Animals , Male , Triiodothyronine/blood , Humans , Mice , Adult , Female , Middle Aged , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Cytokines/blood , Cytokines/metabolism , Thermogenesis/drug effects , Case-Control Studies
7.
Article in English | MEDLINE | ID: mdl-38847150

ABSTRACT

BACKGROUND AND OBJECTIVE: Nitrate, as nitric oxide (NO) donor, has been suggested as a nutrition-based treatment for decreasing the risk of menopause-related obesity. This study aimed to specify the effects of chronic inorganic nitrate administration on uncoupling protein-1 (UCP-1), peroxisome proliferator-activated-receptor-947; (PPAR-947;) coactivator-1945; (PGC-1945;), and PPAR-947; expression in gonadal adipose tissue (GAT) of ovariectomized (OVX) rats. METHODS: Female rats were assigned to 3 groups: Control, OVX, and OVX+nitrate (n=7/group), which consumed water containing inorganic nitrate (100 mg/L) for 9 months. At month 9, GAT was used for the measurement of NO metabolites (NOx), mRNA levels of NO synthases (endothelial (eNOS), inducible (iNOS), neuronal (nNOS)), and mRNA and protein levels of UCP-1, PGC-1945;, and PPAR-947;. RESULTS: OVX rats had lower NOx concentration (45%) and eNOS (38%) and nNOS (30%) expression in GAT that was restored to normal values following nitrate administration. OVX rats had significantly lower mRNA and protein levels of UCP-1 (83% and 30%), PGC-1945; (65% and 39%), and PPAR-947; (66% and 34.5%) in GAT. Chronic inorganic nitrate administration in OVXrats increased mRNA and protein levels of UCP-1 (128% and 34%), PGC-1945; (115% and 43%), and PPAR-947; (236% and 38%), respectively. CONCLUSION: In OVX rats, chronic nitrate administration increased gene and protein levels of UCP-1, PGC-1945;, and PPAR-947; in GAT, indicating the anti-obesity effects of nitrate are partially mediated by the white adipose tissue (WAT) browning. Moreover, the stimulatory effect of inorganic nitrate on the WAT browning in OVX rats was associated with blunting the OVXinduced NO deficiency in GAT.


Subject(s)
Adipose Tissue, Brown , Nitrates , Ovariectomy , Rats, Wistar , Uncoupling Protein 1 , Animals , Female , Nitrates/administration & dosage , Nitrates/metabolism , Rats , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Nitric Oxide/metabolism , Gene Expression Regulation/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
8.
Ann Endocrinol (Paris) ; 85(3): 184-189, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38871497

ABSTRACT

Brown adipose tissue (BAT) and beige adipose tissues are important contributors to cold-induced whole body thermogenesis in rodents. The documentation in humans of cold- and ß-adrenergic receptor agonist-stimulated BAT glucose uptake using positron emission tomography (PET) and of a decrease of this response in individuals with cardiometabolic disorders led to the suggestion that BAT/beige adipose tissues could be relevant targets for prevention and treatment of these conditions. In this brief review, we will critically assess this question by first describing the basic rationale for this affirmation, second by examining the evidence in human studies, and third by discussing the possible means to activate the thermogenic response of these tissues in humans.


Subject(s)
Adipose Tissue, Beige , Adipose Tissue, Brown , Thermogenesis , Humans , Adipose Tissue, Brown/physiology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Thermogenesis/physiology , Adipose Tissue, Beige/metabolism , Adipose Tissue, Beige/physiology , Animals , Positron-Emission Tomography , Adrenergic beta-Agonists/pharmacology , Obesity/metabolism , Obesity/therapy , Cold Temperature
10.
Cell Metab ; 36(6): 1287-1301.e7, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38838641

ABSTRACT

Adipocytes in dermis are considered to be important participants in skin repair and regeneration, but the role of subcutaneous white adipose tissue (sWAT) in skin repair is poorly understood. Here, we revealed the dynamic changes of sWAT during wound healing process. Lineage-tracing mouse studies revealed that sWAT would enter into the large wound bed and participate in the formation of granulation tissue. Moreover, sWAT undergoes beiging after skin injury. Inhibition of sWAT beiging by genetically silencing PRDM16, a key regulator to beiging, hindered wound healing process. The transcriptomics results suggested that beige adipocytes in sWAT abundantly express neuregulin 4 (NRG4), which regulated macrophage polarization and the function of myofibroblasts. In diabetic wounds, the beiging of sWAT was significantly suppressed. Thus, adipocytes from sWAT regulate multiple aspects of repair and may be therapeutic for inflammatory diseases and defective wound healing associated with aging and diabetes.


Subject(s)
Adipose Tissue, White , Skin , Wound Healing , Animals , Adipose Tissue, White/metabolism , Mice , Skin/metabolism , Skin/pathology , Mice, Inbred C57BL , Subcutaneous Fat/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Neuregulins/metabolism , Neuregulins/genetics , Male , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Adipose Tissue, Brown/metabolism , Adipocytes, Beige/metabolism , Macrophages/metabolism , Humans , Myofibroblasts/metabolism
11.
J Nucl Med Technol ; 52(2): 115-120, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839114

ABSTRACT

Brown fat can present challenges in patients with cancer who undergo 18F-FDG PET scans. Uptake of 18F-FDG by brown fat can obscure or appear similar to active oncologic lesions, causing clinical challenges in PET interpretation. Small, retrospective studies have reported environmental and pharmacologic interventions for suppressing brown fat uptake on PET; however, there is no clear consensus on best practices. We sought to characterize practice patterns for strategies to mitigate brown fat uptake of 18F-FDG during PET scanning. Methods: A survey was developed and distributed via e-mail LISTSERV to members of the Children's Oncology Group diagnostic imaging committee, the Society for Nuclear Medicine and Molecular Imaging pediatric imaging council, and the Society of Chiefs of Radiology at Children's Hospitals between April 2022 and February 2023. Responses were stored anonymously in REDCap, aggregated, and summarized using descriptive statistics. Results: Fifty-five complete responses were submitted: 51 (93%) faculty and fellow-level physicians, 2 (4%) technologists, and 2 (4%) respondents not reporting their rank. There were 43 unique institutions represented, including 5 (12%) outside the United States. Thirty-eight of 41 (93%) institutions that responded on environmental interventions reported using warm blankets in the infusion and scanning rooms. Less than a third (n = 13, 30%) of institutions reported use of a pharmacologic intervention, with propranolol (n = 5, 38%) being most common, followed by fentanyl (n = 4, 31%), diazepam (n = 2, 15%), and diazepam plus propranolol (n = 2, 15%). Selection criteria for pharmacologic intervention varied, with the most common criterion being brown fat uptake on a prior scan (n = 6, 45%). Conclusion: Clinical practices to mitigate brown fat uptake on pediatric 18F-FDG PET vary widely. Simple environmental interventions including warm blankets or increasing the temperature of the injection and scanning rooms were not universally reported. Less than a third of institutions use pharmacologic agents for brown fat mitigation.


Subject(s)
Adipose Tissue, Brown , Fluorodeoxyglucose F18 , Hospitals, Pediatric , Positron Emission Tomography Computed Tomography , Humans , Positron Emission Tomography Computed Tomography/methods , Adipose Tissue, Brown/diagnostic imaging , Adipose Tissue, Brown/metabolism , Surveys and Questionnaires , Internationality , Biological Transport , Child
12.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38843936

ABSTRACT

Lipid composition is conserved within sub-cellular compartments to maintain cell function. Lipidomic analyses of liver, muscle, white and brown adipose tissue (BAT) mitochondria revealed substantial differences in their glycerophospholipid (GPL) and free cholesterol (FC) contents. The GPL to FC ratio was 50-fold higher in brown than white adipose tissue mitochondria. Their purity was verified by comparison of proteomes with ER and mitochondria-associated membranes. A lipid signature containing PC and FC, calculated from the lipidomic profiles, allowed differentiation of mitochondria from BAT of mice housed at different temperatures. Elevating FC in BAT mitochondria prevented uncoupling protein (UCP) 1 function, whereas increasing GPL boosted it. Similarly, STARD3 overexpression facilitating mitochondrial FC import inhibited UCP1 function in primary brown adipocytes, whereas a knockdown promoted it. We conclude that the mitochondrial GPL/FC ratio is key for BAT function and propose that targeting it might be a promising strategy to promote UCP1 activity.


Subject(s)
Adipose Tissue, Brown , Cholesterol , Lipidomics , Mitochondria , Uncoupling Protein 1 , Animals , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Mice , Adipose Tissue, Brown/metabolism , Cholesterol/metabolism , Mitochondria/metabolism , Lipidomics/methods , Organ Specificity , Mice, Inbred C57BL , Adipose Tissue, White/metabolism , Glycerophospholipids/metabolism , Male , Lipid Metabolism
13.
Science ; 384(6700): 1111-1117, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843333

ABSTRACT

Brown adipose tissue (BAT) is a heater organ that expresses thermogenic uncoupling protein 1 (UCP1) to maintain high body temperatures during cold stress. BAT thermogenesis is considered an overarching mammalian trait, but its evolutionary origin is unknown. We show that adipose tissue of marsupials, which diverged from eutherian mammals ~150 million years ago, expresses a nonthermogenic UCP1 variant governed by a partial transcriptomic BAT signature similar to that found in eutherian beige adipose tissue. We found that the reconstructed UCP1 sequence of the common eutherian ancestor displayed typical thermogenic activity, whereas therian ancestor UCP1 is nonthermogenic. Thus, mammalian adipose tissue thermogenesis may have evolved in two distinct stages, with a prethermogenic stage in the common therian ancestor linking UCP1 expression to adipose tissue and thermal stress. We propose that in a second stage, UCP1 acquired its thermogenic function specifically in eutherians, such that the onset of mammalian BAT thermogenesis occurred only after the divergence from marsupials.


Subject(s)
Adipose Tissue, Brown , Biological Evolution , Marsupialia , Thermogenesis , Uncoupling Protein 1 , Animals , Humans , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , Eutheria/genetics , Eutheria/physiology , Evolution, Molecular , Marsupialia/genetics , Marsupialia/physiology , Phylogeny , Thermogenesis/genetics , Transcriptome , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
14.
Science ; 384(6700): 1065-1066, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843350

ABSTRACT

Comparative genomics elucidates the steps enabling heat production in fat tissue.


Subject(s)
Adipose Tissue, Brown , Biological Evolution , Mammals , Thermogenesis , Animals , Mammals/genetics , Mammals/physiology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/physiology , Humans , Genomics
15.
Physiol Rep ; 12(11): e16055, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872474

ABSTRACT

This study examined the effects of exercise and detraining at a young age on fat accumulation in various organs. Four-week-old male Otsuka Long-Evans Tokushima Fatty (OLETF) rats were assigned to either the non-exercise sedentary (OLETF Sed) or exercise groups. The exercise group was subdivided into two groups: exercise between 4 and 12 weeks of age (OLETF Ex) and exercise between 4 and 6 weeks of age followed by non-exercise between 6 and 12 weeks of age (OLETF DT). Body weight was significantly lower in the OLETF Ex group than in the OLETF Sed group at 12 weeks of age. Fat accumulation in the epididymal white adipose tissue, liver, and brown adipose tissue was suppressed in the OLETF Ex group. During the exercise period, body weight and food intake in the OLETF DT group were significantly lower than those in the OLETF Sed group. However, food intake was significantly higher in the OLETF DT group than in the OLETF Sed group after exercise cessation, resulting in extreme obesity with fatty liver and brown adipose tissue whitening. Detraining after early-onset exercise promotes hyperphagia, causing extreme obesity. Overeating should be avoided during detraining periods in cases of exercise cessation at a young age.


Subject(s)
Adipose Tissue, Brown , Fatty Liver , Hyperphagia , Obesity , Physical Conditioning, Animal , Rats, Inbred OLETF , Animals , Male , Adipose Tissue, Brown/metabolism , Hyperphagia/physiopathology , Hyperphagia/metabolism , Rats , Fatty Liver/metabolism , Fatty Liver/etiology , Obesity/metabolism , Obesity/physiopathology , Obesity/etiology , Eating , Liver/metabolism , Body Weight
16.
Front Biosci (Landmark Ed) ; 29(6): 208, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38940030

ABSTRACT

Polycystic ovary syndrome (PCOS) is a prevalent reproductive, endocrine, and metabolic disease that affects 5-18% of women worldwide, with a rising incidence. Hyperandrogenemia and insulin resistance are two key pathophysiological factors that contribute to PCOS, both of which contribute to a variety of health issues such as menstrual irregularities, obesity, dysfunctional glucose and lipid homeostasis, infertility, mental disorders, and cardiovascular and cerebrovascular diseases. Despite ongoing studies, the origin and pathogenesis of PCOS remain elusive; there is also a clinical need for simpler, more effective, longer lasting, and more comprehensive treatments for women with PCOS. The gut-fat axis, a critical regulatory route for metabolism, endocrine function, and immune response, has received considerable interest in recent years in the research of the etiology and treatment of metabolic illnesses such as type 2 diabetes mellitus and non-alcoholic fatty liver disease. The latest research in PCOS has revealed significant alterations in the homogeneity and phylogenetic diversity of the gut microbiota. Animal research using fecal microbiota transplantation has confirmed the importance of gut microbiota in regulating insulin sensitivity and sex hormone balance in PCOS. Furthermore, studies have shown a decrease in the volume and/or activity of brown adipose tissue (BAT) in PCOS patients, a change that alters adipokine release, leading to insulin resistance and hyperandrogenemia, aggravating PCOS progression. Given the function of BAT in increasing energy expenditure and alleviating metabolic parameters, efforts to activate BAT or induce browning of white adipose tissue have emerged as possible treatments for PCOS. Recent research has suggested that the gut microbiota can influence BAT creation and activity via metabolites such as short-chain fatty acids and bile acids, as well as the gut-brain axis. Cold exposure, healthy dieting, metformin, bariatric surgery, glucagon-like peptide 1 receptor agonists and melatonin have all been shown in basic and clinical studies to modulate BAT activity by influencing the gut microbiota, demonstrating significant clinical potential. However, more studies into the regulation mechanisms of the gut-BAT axis are required to produce more effective, comfortable, and safe tailored therapeutics for PCOS.


Subject(s)
Adipose Tissue, Brown , Gastrointestinal Microbiome , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/microbiology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/therapy , Polycystic Ovary Syndrome/physiopathology , Humans , Female , Gastrointestinal Microbiome/physiology , Adipose Tissue, Brown/metabolism , Animals , Insulin Resistance , Fecal Microbiota Transplantation , Obesity/microbiology , Obesity/metabolism , Obesity/therapy
17.
Front Biosci (Landmark Ed) ; 29(6): 236, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38940054

ABSTRACT

BACKGROUND: This study aimed to elucidate the molecular mechanism through which C1q/tumor necrosis factor (TNF)-related protein 9 (CTRP9) acts in the formation and differentiation of brown adipose tissue (BAT). METHODS: Adenovirus particles encoding CTRP9 and green fluorescent protein were inoculated into the scapula of C57BL/6J mice and fed a high-fat diet for 8 weeks; the body weight, lipid droplet morphology, glucose tolerance, insulin tolerance, and protein expression levels were analyzed. In addition, CTRP9 adenovirus was transfected into brown preadipocytes, and differentiation was induced to identify the effect of CTRP9 overexpression on adipocyte differentiation. RESULTS: CTRP9 overexpression significantly increased the weight gain of mice. Additionally, the CTRP9 overexpression group exhibited significantly increased adipose tissue weight and glucose clearance rates and decreased insulin sensitivity and serum triglyceride levels compared to the control group. Furthermore, CTRP9 overexpression significantly upregulated the adipose triglyceride lipase (ATGL) and perilipin 1 protein expression levels in BAT. The cell experiment results confirmed that CTRP9 overexpression significantly inhibited the adipogenesis of brown adipocytes as evidenced by the downregulation of uncoupling protein 1, beta-3 adrenergic receptor, ATGL, and hormone-sensitive lipase mRNA levels and the significant suppression of uncoupling protein 1, ATGL, and perilipin 1 protein levels in brown adipocytes. CONCLUSIONS: The finding of this study demonstrated that CTRP9 promotes lipolysis by upregulating ATGL expression in vivo and inhibits the differentiation of brown preadipocytes in vitro.


Subject(s)
Adipose Tissue, Brown , Diet, High-Fat , Lipolysis , Mice, Inbred C57BL , Animals , Diet, High-Fat/adverse effects , Adipose Tissue, Brown/metabolism , Male , Mice , Adiponectin/metabolism , Adiponectin/genetics , Insulin Resistance , Lipase/metabolism , Lipase/genetics , Cell Differentiation , Adipogenesis/genetics , Perilipin-1/metabolism , Perilipin-1/genetics , Acyltransferases , Glycoproteins
18.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892320

ABSTRACT

Declining estrogen (E2) leads to physical inactivity and adipose tissue (AT) dysfunction. Mechanisms are not fully understood, but E2's effects on dopamine (DA) activity in the nucleus accumbens (NAc) brain region may mediate changes in mood and voluntary physical activity (PA). Our prior work revealed that loss of E2 robustly affected NAc DA-related gene expression, and the pattern correlated with sedentary behavior and visceral fat. The current study used a new transgenic mouse model (D1ERKO) to determine whether the abolishment of E2 receptor alpha (ERα) signaling within DA-rich brain regions affects PA and AT metabolism. Adult male and female wild-type (WT) and D1ERKO (KD) mice were assessed for body composition, energy intake (EE), spontaneous PA (SPA), and energy expenditure (EE); underwent glucose tolerance testing; and were assessed for blood biochemistry. Perigonadal white AT (PGAT), brown AT (BAT), and NAc brain regions were assessed for genes and proteins associated with DA, E2 signaling, and metabolism; AT sections were also assessed for uncoupling protein (UCP1). KD mice had greater lean mass and EE (genotype effects) and a visible change in BAT phenotype characterized by increased UCP1 staining and lipid depletion, an effect seen only among females. Female KD had higher NAc Oprm1 transcript levels and greater PGAT UCP1. This group tended to have improved glucose tolerance (p = 0.07). NAc suppression of Esr1 does not appear to affect PA, yet it may directly affect metabolism. This work may lead to novel targets to improve metabolic dysfunction following E2 loss, possibly by targeting the NAc.


Subject(s)
Adipose Tissue , Energy Metabolism , Estrogen Receptor alpha , Nucleus Accumbens , Receptors, Dopamine D1 , Animals , Nucleus Accumbens/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Mice , Female , Male , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/genetics , Energy Metabolism/genetics , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Mice, Knockout , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Brain/metabolism , Mice, Transgenic , Gene Knockdown Techniques , Mice, Inbred C57BL
19.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38836615

ABSTRACT

About half of the world population carries at least one allele of the Ala92-DIO2, which slows down the activity of the type 2 deiodinase (D2), the enzyme that activates T4 to T3. Carrying the Ala92-DIO2 allele has been associated with increased body mass index and insulin resistance, but this has not been reproduced in all populations. To test if the genetic background affects the impact of this polymorphism, here we studied the genetically distant C57Bl/6J (B6) and FVB/N (FVB) mice carrying the Ala92-Dio2 allele as compared to control mice carrying the Thr92-Dio2 allele. Whereas B6-Ala92-Dio2 and B6-Thr92-Dio2 mice-fed chow or high-fat diet-behaved metabolically similar in studies using indirect calorimetry, glucose- and insulin tolerance tests, and measuring white adipose tissue (WAT) weight and liver steatosis, major differences were observed between FVB-Ala92-Dio2 and FVB-Thr92-Dio2 mice: carrying the Ala92-Dio2 allele (on a chow diet) resulted in hypercholesterolemia, smaller WAT pads, hepatomegaly, steatosis, and transcriptome changes in the interscapular brown adipose tissue (iBAT) typical of ER stress and apoptosis. Acclimatization at thermoneutrality (30 °C) eliminated most of the metabolic phenotype, indicating that impaired adaptive (BAT) thermogenesis can be involved. In conclusion, the metabolic impact of carrying the Ala92-Dio2 allele depends greatly on the genetic background of the mouse, varying from no phenotype in B6 mice to a major phenotype in FVB mice. These results will help the planning of future clinical trials studying the Thr92Ala-DIO2 polymorphism and may explain why some clinical studies performed in different populations across the globe have obtained inconsistent results.


Subject(s)
Iodide Peroxidase , Iodothyronine Deiodinase Type II , Mice, Inbred C57BL , Animals , Male , Iodide Peroxidase/genetics , Mice , Diet, High-Fat , Genetic Background , Adipose Tissue, White/metabolism , Adipose Tissue, Brown/metabolism , Polymorphism, Genetic , Insulin Resistance/genetics , Fatty Liver/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...