Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.521
Filter
1.
FASEB J ; 38(11): e23717, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38837270

ABSTRACT

Selenoprotein I (Selenoi) is highly expressed in liver and plays a key role in lipid metabolism as a phosphatidylethanolamine (PE) synthase. However, the precise function of Selenoi in the liver remains elusive. In the study, we generated hepatocyte-specific Selenoi conditional knockout (cKO) mice on a high-fat diet to identify the physiological function of Selenoi. The cKO group exhibited a significant increase in body weight, with a 15.6% and 13.7% increase in fat accumulation in white adipose tissue (WAT) and the liver, respectively. Downregulation of the lipolysis-related protein (p-Hsl) and upregulation of the adipogenesis-related protein (Fasn) were observed in the liver of cKO mice. The cKO group also showed decreased oxygen consumption (VO2), carbon dioxide production (VCO2), and energy expenditure (p < .05). Moreover, various metabolites of the steroid hormone synthesis pathway were affected in the liver of cKO mice. A potential cascade of Selenoi-phosphatidylethanolamine-steroid hormone synthesis might serve as a core mechanism that links hepatocyte-specific Selenoi cKO to biochemical and molecular reactions. In conclusion, we revealed that Selenoi inhibits body fat accumulation and hepatic steatosis and elevates energy consumption; this protein could also be considered a therapeutic target for such related diseases.


Subject(s)
Fatty Liver , Hepatocytes , Mice, Knockout , Obesity , Animals , Mice , Obesity/metabolism , Obesity/genetics , Obesity/etiology , Hepatocytes/metabolism , Fatty Liver/metabolism , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/pathology , Selenoproteins/metabolism , Selenoproteins/genetics , Diet, High-Fat/adverse effects , Male , Liver/metabolism , Energy Metabolism , Lipid Metabolism , Mice, Inbred C57BL , Adipose Tissue, White/metabolism
2.
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
3.
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
4.
Cells ; 13(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38727299

ABSTRACT

The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.


Subject(s)
Adipogenesis , Adipose Tissue, White , Aging , Obesity , Humans , Aging/pathology , Obesity/pathology , Obesity/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Adipocytes/metabolism , Adipocytes/pathology
5.
Sci Signal ; 17(836): eadq3321, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38743810

ABSTRACT

Activation of GPR81 in white adipose tissue by lactate results in cancer-associated cachexia.


Subject(s)
Cachexia , Lactic Acid , Neoplasms , Receptors, G-Protein-Coupled , Humans , Lactic Acid/metabolism , Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Cachexia/metabolism , Animals , Adipose Tissue, White/metabolism
6.
J Diabetes Res ; 2024: 5511454, 2024.
Article in English | MEDLINE | ID: mdl-38736904

ABSTRACT

Adipose tissue dysfunction is seen among obese and type 2 diabetic individuals. Adipocyte proliferation and hypertrophy are the root causes of adipose tissue expansion. Solute carrier family 25 member 28 (SLC25A28) is an iron transporter in the inner mitochondrial membrane. This study is aimed at validating the involvement of SLC25A28 in adipose accumulation by tail vein injection of adenovirus (Ad)-SLC25A28 and Ad-green fluorescent protein viral particles into C57BL/6J mice. After 16 weeks, the body weight of the mice was measured. Subsequently, morphological analysis was performed to establish a high-fat diet (HFD)-induced model. SLC25A28 overexpression accelerated lipid accumulation in white and brown adipose tissue (BAT), enhanced body weight, reduced serum triglyceride (TG), and impaired serum glucose tolerance. The protein expression level of lipogenesis, lipolysis, and serum adipose secretion hormone was evaluated by western blotting. The results showed that adipose TG lipase (ATGL) protein expression was reduced significantly in white and BAT after overexpression SLC25A28 compared to the control group. Moreover, SLC25A28 overexpression inhibited the BAT formation by downregulating UCP-1 and the mitochondrial biosynthesis marker PGC-1α. Serum adiponectin protein expression was unregulated, which was consistent with the expression in inguinal white adipose tissue (iWAT). Remarkably, serum fibroblast growth factor (FGF21) protein expression was negatively related to the expansion of adipose tissue after administrated by Ad-SLC25A28. Data from the current study indicate that SLC25A28 overexpression promotes diet-induced obesity and accelerates lipid accumulation by regulating hormone secretion and inhibiting lipolysis in adipose tissue.


Subject(s)
Adipogenesis , Adipose Tissue, Brown , Adipose Tissue, White , Diet, High-Fat , Lipase , Mice, Inbred C57BL , Animals , Mice , Male , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Lipase/metabolism , Lipase/genetics , Obesity/metabolism , Lipolysis , Uncoupling Protein 1/metabolism , Fibroblast Growth Factors/metabolism , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Adipocytes/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Lipogenesis , Acyltransferases
7.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731880

ABSTRACT

Adipose tissue is a multifunctional organ that regulates many physiological processes such as energy homeostasis, nutrition, the regulation of insulin sensitivity, body temperature, and immune response. In this review, we highlight the relevance of the different mediators that control adipose tissue activity through a systematic review of the main players present in white and brown adipose tissues. Among them, inflammatory mediators secreted by the adipose tissue, such as classical adipokines and more recent ones, elements of the immune system infiltrated into the adipose tissue (certain cell types and interleukins), as well as the role of intestinal microbiota and derived metabolites, have been reviewed. Furthermore, anti-obesity mediators that promote the activation of beige adipose tissue, e.g., myokines, thyroid hormones, amino acids, and both long and micro RNAs, are exhaustively examined. Finally, we also analyze therapeutic strategies based on those mediators that have been described to date. In conclusion, novel regulators of obesity, such as microRNAs or microbiota, are being characterized and are promising tools to treat obesity in the future.


Subject(s)
Adipose Tissue , Obesity , Humans , Animals , Obesity/metabolism , Adipose Tissue/metabolism , Adipokines/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Gastrointestinal Microbiome , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Inflammation Mediators/metabolism , Energy Metabolism
8.
Nat Metab ; 6(5): 963-979, 2024 May.
Article in English | MEDLINE | ID: mdl-38693320

ABSTRACT

Subcutaneous white adipose tissue (scWAT) is a dynamic storage and secretory organ that regulates systemic homeostasis, yet the impact of endurance exercise training (ExT) and sex on its molecular landscape is not fully established. Utilizing an integrative multi-omics approach, and leveraging data generated by the Molecular Transducers of Physical Activity Consortium (MoTrPAC), we show profound sexual dimorphism in the scWAT of sedentary rats and in the dynamic response of this tissue to ExT. Specifically, the scWAT of sedentary females displays -omic signatures related to insulin signaling and adipogenesis, whereas the scWAT of sedentary males is enriched in terms related to aerobic metabolism. These sex-specific -omic signatures are preserved or amplified with ExT. Integration of multi-omic analyses with phenotypic measures identifies molecular hubs predicted to drive sexually distinct responses to training. Overall, this study underscores the powerful impact of sex on adipose tissue biology and provides a rich resource to investigate the scWAT response to ExT.


Subject(s)
Adipose Tissue, White , Physical Conditioning, Animal , Sex Characteristics , Subcutaneous Fat , Animals , Male , Female , Rats , Adipose Tissue, White/metabolism , Subcutaneous Fat/metabolism , Adipogenesis , Rats, Sprague-Dawley , Multiomics
9.
Biochem Biophys Res Commun ; 716: 149998, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38692012

ABSTRACT

The equilibrium between the hypertrophic growth of existing adipocytes and adipogenesis is vital in managing metabolic stability in white adipocytes when faced with overnutrition. Adipogenesis has been established as a key player in combating metabolic irregularities caused by various factors. However, the benefits of increasing adipogenesis-mediated white adipose tissue (WAT) expansion for metabolic health regulation remain uncertain. Our findings reveal an increase in Impdh2 expression during the adipogenesis phase, both in vivo and in vitro. Xmp enhances adipogenic potential by fostering mitotic clonal expansion (MCE). The conditional knockout of Impdh2 in adipocyte progenitor cells(APCs) in adult and aged mice effectively curbs white adipose tissue expansion, ameliorates glucose tolerance, and augments energy expenditure under high-fat diet (HFD). However, no significant difference is observed under normal chow diet (NCD). Concurrently, the knockout of Impdh2 in APCs significantly reduces the count of new adipocytes induced by HFD, without affecting adipocyte size. Mechanistically, Impdh2 regulates the proliferation of APCs during the MCE phase via Xmp. Exogenous Xmp can significantly offset the reduction in adipogenic abilities of APCs due to Impdh2 deficiency. In summary, we discovered that adipogenesis-mediated WAT expansion, induced by overnutrition, also contributes to metabolic abnormalities. Moreover, the pivotal role of Impdh2 in regulating adipogenesis in APCs offers a novel therapeutic approach to combat obesity.


Subject(s)
Adipocytes , Adipogenesis , Adipose Tissue, White , Diet, High-Fat , Mice, Knockout , Overnutrition , Animals , Adipose Tissue, White/metabolism , Adipogenesis/genetics , Overnutrition/metabolism , Overnutrition/genetics , Mice , Adipocytes/metabolism , Mice, Inbred C57BL , Male , Energy Metabolism/genetics , Gene Deletion , Cell Proliferation , Stem Cells/metabolism , Stem Cells/cytology , Stem Cells/pathology
10.
Front Endocrinol (Lausanne) ; 15: 1385811, 2024.
Article in English | MEDLINE | ID: mdl-38765953

ABSTRACT

Background: Thermogenic beige adipocytes, which dissipate energy as heat, are found in neonates and adults. Recent studies show that neonatal beige adipocytes are highly plastic and contribute to >50% of beige adipocytes in adults. Neonatal beige adipocytes are distinct from recruited beige adipocytes in that they develop independently of temperature and sympathetic innervation through poorly defined mechanisms. Methods: We characterized the neonatal beige adipocytes in the inguinal white adipose tissue (iWAT) of C57BL6 postnatal day 3 and 20 mice (P3 and P20) by imaging, genome-wide RNA-seq analysis, ChIP-seq analysis, qRT-PCR validation, and biochemical assays. Results: We found an increase in acetylated histone 3 lysine 27 (H3K27ac) on the promoter and enhancer regions of beige-specific gene UCP1 in iWAT of P20 mice. Furthermore, H3K27ac ChIP-seq analysis in the iWAT of P3 and P20 mice revealed strong H3K27ac signals at beige adipocyte-associated genes in the iWAT of P20 mice. The integration of H3K27ac ChIP-seq and RNA-seq analysis in the iWAT of P20 mice reveal epigenetically active signatures of beige adipocytes, including oxidative phosphorylation and mitochondrial metabolism. We identify the enrichment of GA-binding protein alpha (GABPα) binding regions in the epigenetically active chromatin regions of the P20 iWAT, particularly on beige genes, and demonstrate that GABPα is required for beige adipocyte differentiation. Moreover, transcriptomic analysis and glucose oxidation assays revealed increased glycolytic activity in the neonatal iWAT from P20. Conclusions: Our findings demonstrate that epigenetic mechanisms regulate the development of peri-weaning beige adipocytes via GABPα. Further studies to better understand the upstream mechanisms that regulate epigenetic activation of GABPα and characterization of the metabolic identity of neonatal beige adipocytes will help us harness their therapeutic potential in metabolic diseases.


Subject(s)
Adipocytes, Beige , Adipogenesis , Adipose Tissue, White , Animals, Newborn , Chromatin , Epigenesis, Genetic , GA-Binding Protein Transcription Factor , Mice, Inbred C57BL , Animals , Mice , Adipocytes, Beige/metabolism , Chromatin/metabolism , Chromatin/genetics , Adipogenesis/genetics , Adipose Tissue, White/metabolism , GA-Binding Protein Transcription Factor/metabolism , GA-Binding Protein Transcription Factor/genetics , Male , Thermogenesis/genetics , Histones/metabolism , Histones/genetics
11.
Sci Rep ; 14(1): 12530, 2024 05 31.
Article in English | MEDLINE | ID: mdl-38822155

ABSTRACT

Growing obesity is linked to shifts in dietary patterns, particularly the increased intake of ultra-processed high-fat foods. This study aimed to evaluate the effects of interesterified palm oil consumption on glucose homeostasis, adipose tissue remodeling, and hepatic lipogenesis in C57BL/6 mice fed a high-fat diet. Sixty C57BL/6 mice were divided into four groups (n = 15): the control group (C) fed a standard diet (4% soybean oil), the high-fat group (HF) (23.8% lard), the high palm oil fat group (HFP) (23.8% palm oil), and the high interesterified palm fat group (HFI) (23.8% interesterified palm oil) for 8 weeks (all groups received 50% energy from lipids). The HFI group exhibited higher body mass than the HF group (+ 11%, P < 0.05), which was attributed to an increased percentage of fat mass. Plasma concentrations of IL-6, insulin, and HOMA-IR were also elevated in the HFI group. Both the HFP and HFI groups showed hypertrophied adipocytes and pancreatic islets, increased alpha and beta cell masses, hepatic steatosis, low expression of genes related to beta-oxidation, and upregulated lipogenesis. In conclusion, the consumption of interesterified palm oil alters inflammatory and glucose profiles.


Subject(s)
Adipose Tissue, White , Diet, High-Fat , Inflammation , Mice, Inbred C57BL , Palm Oil , Animals , Diet, High-Fat/adverse effects , Mice , Inflammation/metabolism , Inflammation/chemically induced , Inflammation/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Male , Lipogenesis/drug effects , Obesity/metabolism , Obesity/etiology , Obesity/chemically induced , Liver/metabolism , Liver/drug effects , Liver/pathology , Insulin/blood , Insulin/metabolism , Insulin Resistance
12.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167227, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733774

ABSTRACT

Olanzapine (OLA) is a highly obesogenic second-generation antipsychotic (SGA). Recently we demonstrated that, contrarily to OLA oral treatment, intraperitoneal (i.p.) administration resulted in weight loss and absence of hepatic steatosis in wild-type (WT) and protein tyrosine phosphatase 1B (PTP1B)-deficient (KO) male mice. This protection relied on two central-peripheral axes connecting hypothalamic AMPK with brown/inguinal white adipose tissue (BAT/iWAT) uncoupling protein-1 (UCP-1) and hypothalamic JNK with hepatic fatty acid synthase (FAS). Herein, we addressed OLA i.p. treatment effects in WT and PTP1B-KO female mice. Contrarily to our previous results in WT females receiving OLA orally, the i.p. treatment did not induce weight gain or hyperphagia. Molecularly, in females OLA failed to diminish hypothalamic phospho-AMPK or elevate BAT UCP-1 and energy expenditure (EE) despite the preservation of iWAT browning. Conversely, OLA i.p. treatment in ovariectomized mice reduced hypothalamic phospho-AMPK, increased BAT/iWAT UCP-1 and EE, and induced weight loss as occurred in males. Pretreatment of hypothalamic neurons with 17ß-estradiol (E2) abolished OLA effects on AMPK. Moreover, neither hypothalamic JNK activation nor hepatic FAS upregulation were found in WT and PTP1B-KO females receiving OLA via i.p. Importantly, this axis was reestablished upon ovariectomy. In this line, E2 prevented OLA-induced phospho-JNK in hypothalamic neurons. These results support the role of estrogens in sex-related dimorphism in OLA treatment. This study evidenced the benefit of OLA i.p. administration in preventing its obesogenic effects in female mice that could offer clinical value.


Subject(s)
Adipose Tissue, Brown , Estrogens , Hypothalamus , Liver , Mice, Knockout , Olanzapine , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Uncoupling Protein 1 , Animals , Female , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Hypothalamus/metabolism , Hypothalamus/drug effects , Mice , Liver/metabolism , Liver/drug effects , Estrogens/metabolism , Estrogens/pharmacology , Olanzapine/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Male , Energy Metabolism/drug effects , Injections, Intraperitoneal , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Mice, Inbred C57BL , Estradiol/pharmacology , Ovariectomy
13.
Lipids Health Dis ; 23(1): 144, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760797

ABSTRACT

BACKGROUND: Cancer-associated cachexia (CAC) arises from malignant tumors and leads to a debilitating wasting syndrome. In the pathophysiology of CAC, the depletion of fat plays an important role. The mechanisms of CAC-induced fat loss include the enhancement of lipolysis, inhibition of lipogenesis, and browning of white adipose tissue (WAT). However, few lipid-metabolic enzymes have been reported to be involved in CAC. This study hypothesized that ELOVL6, a critical enzyme for the elongation of fatty acids, may be involved in fat loss in CAC. METHODS: Transcriptome sequencing technology was used to identify CAC-related genes in the WAT of a CAC rodent model. Then, the expression level of ELOVL6 and the fatty acid composition were analyzed in a large clinical sample. Elovl6 was knocked down by siRNA in 3T3-L1 mouse preadipocytes to compare with wild-type 3T3-L1 cells treated with tumor cell conditioned medium. RESULTS: In the WAT of patients with CAC, a significant decrease in the expression of ELOVL6 was found, which was linearly correlated with the extent of body mass reduction. Gas chromatographic analysis revealed an increase in palmitic acid (C16:0) and a decrease in linoleic acid (C18:2n-6) in these tissue samples. After treatment with tumor cell-conditioned medium, 3T3-L1 mouse preadipocytes showed a decrease in Elovl6 expression, and Elovl6-knockdown cells exhibited a reduction in preadipocyte differentiation and lipogenesis. Similarly, the knockdown of Elovl6 in 3T3-L1 cells resulted in a significant increase in palmitic acid (C16:0) and a marked decrease in oleic acid (C18:1n-9) content. CONCLUSION: Overall, the expression of ELOVL6 was decreased in the WAT of CAC patients. Decreased expression of ELOVL6 might induce fat loss in CAC patients by potentially altering the fatty acid composition of adipocytes. These findings suggest that ELOVL6 may be used as a valuable biomarker for the early diagnosis of CAC and may hold promise as a target for future therapies.


Subject(s)
3T3-L1 Cells , Adipose Tissue, White , Cachexia , Fatty Acid Elongases , Neoplasms , Fatty Acid Elongases/genetics , Fatty Acid Elongases/metabolism , Animals , Cachexia/genetics , Cachexia/metabolism , Cachexia/pathology , Mice , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/complications , Neoplasms/pathology , Male , Female , Palmitic Acid/metabolism , Lipogenesis/genetics , Middle Aged , Fatty Acids/metabolism
14.
Medicina (Kaunas) ; 60(5)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38792922

ABSTRACT

Background and Objectives: The hormonal state of hypoestrogenism is associated with the accumulation of white adipose tissue, which can induce an increase in pro-inflammatory markers, leading to progressive health complications. Melatonin can act on adipose tissue mass, promoting its reduction and influencing inflammation, reducing IL-6 and releasing IL-10, pro- and anti-inflammatory markers, respectively. However, the role of melatonin regarding such parameters under the context of hypoestrogenism remains unknown. The aim of this study was to determine the effect of 12 weeks of hypoestrogenism and melatonin on white adipose tissue mass and circulating levels of IL-6, IL-10, TGF-ß-1, and leukotriene C4 (LTC4). Materials and Methods: The animals (Wistar rats with sixteen weeks of age at the beginning of the experiment) under hypoestrogenism were submitted to the surgical technique of bilateral ovariectomy. The animals received melatonin (10 mg·kg-1) or vehicles by orogastric gavage every day for 12 weeks and administration occurred systematically 1 h after the beginning of the dark period. White adipose tissue (perigonadal, peritoneal, and subcutaneous) was collected for mass recording, while blood was collected for the serum determination of IL-6, IL-10, TGF-ß-1, and LTC4. Results: Hypoestrogenism increased the perigonadal and subcutaneous mass and IL-6 levels. Melatonin kept hypoestrogenic animals in physiological conditions similar to the control group and increased thymus tissue mass. Conclusions: Hypoestrogenism appears to have a negative impact on white adipose tissue mass and IL-6 and although melatonin commonly exerts a significant effect in preventing these changes, this study did not have a sufficiently negative impact caused by hypoestrogenism for melatonin to promote certain benefits.


Subject(s)
Interleukin-6 , Melatonin , Rats, Wistar , Animals , Melatonin/analysis , Melatonin/blood , Rats , Female , Interleukin-6/blood , Interleukin-6/analysis , Biomarkers/blood , Biomarkers/analysis , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Interleukin-10/blood , Ovariectomy , Inflammation , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/analysis , Estrogens/blood , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism
15.
Front Endocrinol (Lausanne) ; 15: 1344262, 2024.
Article in English | MEDLINE | ID: mdl-38559696

ABSTRACT

Obesity, a multifactorial disease with many complications, has become a global epidemic. Weight management, including dietary supplementation, has been confirmed to provide relevant health benefits. However, experimental evidence and mechanistic elucidation of dietary supplements in this regard are limited. Here, the weight loss efficacy of MHP, a commercial solid beverage consisting of mulberry leaf aqueous extract and Hippophae protein peptides, was evaluated in a high-fat high-fructose (HFF) diet-induced rat model of obesity. Body component analysis and histopathologic examination confirmed that MHP was effective to facilitate weight loss and adiposity decrease. Pathway enrichment analysis with differential metabolites generated by serum metabolomic profiling suggests that PPAR signal pathway was significantly altered when the rats were challenged by HFF diet but it was rectified after MHP intervention. RNA-Seq based transcriptome data also indicates that MHP intervention rectified the alterations of white adipose tissue mRNA expressions in HFF-induced obese rats. Integrated omics reveals that the efficacy of MHP against obesogenic adipogenesis was potentially associated with its regulation of PPARγ and FGFR1 signaling pathway. Collectively, our findings suggest that MHP could improve obesity, providing an insight into the use of MHP in body weight management.


Subject(s)
Hippophae , Morus , Rats , Animals , PPAR gamma/genetics , PPAR gamma/metabolism , Hippophae/metabolism , Morus/metabolism , Diet, High-Fat/adverse effects , Obesity/metabolism , Adipose Tissue, White/metabolism , Signal Transduction , Weight Loss
16.
PeerJ ; 12: e17105, 2024.
Article in English | MEDLINE | ID: mdl-38563016

ABSTRACT

Background: Colorectal cancer (CRC) is one of the most common cancers worldwide and is related to diet and obesity. Currently, crosstalk between lipid metabolism and CRC has been reported; however, the specific mechanism is not yet understood. In this study, we screened differentially expressed long non-coding RNAs (lncRNAs) and mRNAs from primary cancer, paracancer, and white adipose tissue of CRC patients. We screened and analyzed the genes differentially expressed between primary and paracancer tissue and between paracancer and white adipose tissue but not between primary and white adipose tissue. According to the results of the biological analysis, we speculated a lncRNA (MIR503HG) that may be involved in the crosstalk between CRC and lipid metabolism through exosome delivery. Methods: We screened differentially expressed long non-coding RNAs (lncRNAs) and mRNAs from primary cancer, paracancer, and white adipose tissue of CRC patients. We screened and analyzed the genes differentially expressed between primary and paracancer tissue and between paracancer and white adipose tissue but not between primary and white adipose tissue. Results: We speculated a lncRNA (MIR503HG) that may be involved in the crosstalk between CRC and lipid metabolism through exosome delivery. Conclusions: In this study, the findings raise the possibility of crosstalk between lipid metabolism and CRC through the exosomal delivery of lncRNAs.


Subject(s)
Colorectal Neoplasms , RNA, Long Noncoding , Humans , Transcriptome/genetics , Gene Expression Profiling/methods , RNA, Long Noncoding/genetics , Adipose Tissue, White/metabolism , Colorectal Neoplasms/genetics , RNA, Messenger/genetics
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(3): 499-506, 2024 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-38597441

ABSTRACT

OBJECTIVE: To investigate the effects of α7 nicotinic acetylcholine receptor (nAChR) agonist on ß3-adrenoceptor agonist-induced impairment of white fat homeostasis and beige adipose formation and heat production in obese mice. METHODS: Forty obese C57BL/6J mice were randomized into high-fat feeding group, ß3-adrenoceptor agonist-treated model group, α7 nAChR agonist group, and α7 nAChR inhibitor group (n=10), with another 10 mice with normal feeding as the blank control group. White adipose tissue from the epididymis of the mice were sampled for HE staining of the adipocytes. The expression levels of TNF-α, IL-1ß, IL-10 and TGF-ß in the white adipose tissue were determined by ELISA, and the mRNA levels of iNOS, Arg1, UCP-1, PRDM-16 and PGC-1α were detected using RT-qPCR. Western blotting was performed to detect the expression levels of NF-κB P65, p-JAK2, p-STAT3 in the white adipose tissue. RESULTS: Compared with those in the blank control group, the mice with high-fat feeding showed significantly increased body weight, more fat vacuoles in the white adipose tissue, increased volume of lipid droplets in the adipocytes, upregulated iNOS mRNA expression and protein expression of TNF-α and IL-1ß, and lowered expression of Arg-1 mRNA and IL-10 and TGF-ß proteins (P < 0.01). Treatment with α7 nAChR significantly reduced mRNA levels of PRDM-16, PGC-1α and UCP-1, lowered TNF-α and IL-1ß expressions, increased IL-10 and TGF-ß expressions, and reduced M1/M2 macrophage ratio in the white adipose tissues (P < 0.05 or 0.01). CONCLUSION: Activation of α7 nAchR improves white adipose tissue homeostasis impairment induced by ß3 agonist, promotes transformation of M1 to M2 macrophages, reduces inflammatory response in white adipose tissue, and promote beige adipogenesis and thermogenesis in obese mice.


Subject(s)
Interleukin-10 , alpha7 Nicotinic Acetylcholine Receptor , Animals , Male , Mice , Adipogenesis , Adipose Tissue, White/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Homeostasis , Mice, Inbred C57BL , Mice, Obese , Receptors, Adrenergic/metabolism , RNA, Messenger/metabolism , Thermogenesis , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Proc Natl Acad Sci U S A ; 121(16): e2318935121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38588421

ABSTRACT

Glucose is required for generating heat during cold-induced nonshivering thermogenesis in adipose tissue, but the regulatory mechanism is largely unknown. CREBZF has emerged as a critical mechanism for metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease (NAFLD). We investigated the roles of CREBZF in the control of thermogenesis and energy metabolism. Glucose induces CREBZF in human white adipose tissue (WAT) and inguinal WAT (iWAT) in mice. Lys208 acetylation modulated by transacetylase CREB-binding protein/p300 and deacetylase HDAC3 is required for glucose-induced reduction of proteasomal degradation and augmentation of protein stability of CREBZF. Glucose induces rectal temperature and thermogenesis in white adipose of control mice, which is further potentiated in adipose-specific CREBZF knockout (CREBZF FKO) mice. During cold exposure, CREBZF FKO mice display enhanced thermogenic gene expression, browning of iWAT, and adaptive thermogenesis. CREBZF associates with PGC-1α to repress thermogenic gene expression. Expression levels of CREBZF are negatively correlated with UCP1 in human adipose tissues and increased in WAT of obese ob/ob mice, which may underscore the potential role of CREBZF in the development of compromised thermogenic capability under hyperglycemic conditions. Our results reveal an important mechanism of glucose sensing and thermogenic inactivation through reversible acetylation.


Subject(s)
Adipose Tissue, Brown , Glucose , Mice , Humans , Animals , Glucose/metabolism , Adipose Tissue, Brown/metabolism , Acetylation , Adipose Tissue, White/metabolism , Energy Metabolism , Obesity/genetics , Obesity/metabolism , Thermogenesis/genetics , Mice, Inbred C57BL , Basic-Leucine Zipper Transcription Factors/metabolism
19.
Life Sci ; 345: 122607, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38583857

ABSTRACT

Diabetes mellitus is a disorder characterised metabolic dysfunction that results in elevated glucose level in the bloodstream. Diabetes is of two types, type1 and type 2 diabetes. Obesity is considered as one of the major reasons intended for incidence of diabetes hence it turns out to be essential to study about the adipose tissue which is responsible for fat storage in body. Adipose tissues play significant role in maintaining the balance between energy stabilization and homeostasis. The three forms of adipose tissue are - White adipose tissue (WAT), Brown adipose tissue (BAT) and Beige adipose tissue (intermediate form). The amount of BAT gets reduced, and WAT starts to increase with the age. WAT when exposed to certain stimuli gets converted to BAT by the help of certain transcriptional regulators. The browning of WAT has been a matter of study to treat the metabolic disorders and to initiate the expenditure of energy. The three main regulators responsible for the browning of WAT are PRDM16, PPARγ and PGC-1α via various cellular and molecular mechanism. Presented review article includes the detailed elaborative aspect of genes and proteins involved in conversion of WAT to BAT.


Subject(s)
Adipose Tissue, Brown , Diabetes Mellitus, Type 2 , Humans , Adipose Tissue, Brown/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Adiposity , Transcription Factors/metabolism , Adipose Tissue, White/metabolism , Thermogenesis/genetics
20.
Food Funct ; 15(8): 4515-4526, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38567805

ABSTRACT

Guanidinoacetic acid (GAA) is a naturally occurring amino acid derivative that plays a critical role in energy metabolism. In recent years, a growing body of evidence has emerged supporting the importance of GAA in metabolic dysfunction. Hence, we aimed to investigate the effects of GAA on hepatic and adipose tissue metabolism, as well as systemic inflammatory responses in obese middle-aged mice models and attempted to explore the underlying mechanism. We found that dietary supplementation of GAA inhibited inguinal white adipose tissue (iWAT) hypertrophy in high-fat diet (HFD)-fed mice. In addition, GAA supplementation observably decreased the levels of some systemic inflammatory factors, including IL-4, TNF-α, IL-1ß, and IL-6. Intriguingly, GAA supplementation ameliorated hepatic steatosis and lipid deposition in HFD-fed mice, which was revealed by decreased levels of TG, TC, LDL-C, PPARγ, SREBP-1c, FASN, ACC, FABP1, and APOB and increased levels of HDL-C in the liver. Moreover, GAA supplementation increased the expression of browning markers and mitochondrial-related genes in the iWAT. Further investigation showed that dietary GAA promoted the browning of the iWAT via activating the AMPK/Sirt1 signaling pathway and might be associated with futile creatine cycling in obese mice. These results indicate that GAA has the potential to be used as an effective ingredient in dietary interventions and thus may play an important role in ameliorating and preventing HFD-induced obesity and related metabolic diseases.


Subject(s)
Adipose Tissue, Brown , Adipose Tissue, White , Diet, High-Fat , Glycine , Glycine/analogs & derivatives , Inflammation , Mice, Inbred C57BL , Obesity , Animals , Mice , Diet, High-Fat/adverse effects , Male , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Obesity/metabolism , Obesity/drug therapy , Glycine/pharmacology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Inflammation/drug therapy , Fatty Liver/drug therapy , Fatty Liver/metabolism , Liver/metabolism , Liver/drug effects , Dietary Supplements
SELECTION OF CITATIONS
SEARCH DETAIL
...