Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nature ; 610(7932): 562-568, 2022 10.
Article in English | MEDLINE | ID: mdl-36261549

ABSTRACT

Tobacco smoking is positively correlated with non-alcoholic fatty liver disease (NAFLD)1-5, but the underlying mechanism for this association is unclear. Here we report that nicotine accumulates in the intestine during tobacco smoking and activates intestinal AMPKα. We identify the gut bacterium Bacteroides xylanisolvens as an effective nicotine degrader. Colonization of B. xylanisolvens reduces intestinal nicotine concentrations in nicotine-exposed mice, and it improves nicotine-exacerbated NAFLD progression. Mechanistically, AMPKα promotes the phosphorylation of sphingomyelin phosphodiesterase 3 (SMPD3), stabilizing the latter and therefore increasing intestinal ceramide formation, which contributes to NAFLD progression to non-alcoholic steatohepatitis (NASH). Our results establish a role for intestinal nicotine accumulation in NAFLD progression and reveal an endogenous bacterium in the human intestine with the ability to metabolize nicotine. These findings suggest a possible route to reduce tobacco smoking-exacerbated NAFLD progression.


Subject(s)
Bacteria , Intestines , Nicotine , Non-alcoholic Fatty Liver Disease , Tobacco Smoking , Animals , Humans , Mice , Bacteria/drug effects , Bacteria/metabolism , Ceramides/biosynthesis , Nicotine/adverse effects , Nicotine/metabolism , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/microbiology , Sphingomyelin Phosphodiesterase/metabolism , Tobacco Smoking/adverse effects , Tobacco Smoking/metabolism , Intestines/drug effects , Intestines/microbiology , AMP-Activated Protein Kinases/metabolism , Disease Progression
2.
J Gastroenterol Hepatol ; 39(6): 1134-1144, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615196

ABSTRACT

BACKGROUND AND AIM: Small heterodimer partner (SHP, encoded by NR0B2) plays an important role in maintaining bile acid homeostasis. The loss of the hepatic farnesoid X receptor (FXR)/SHP signal can cause severe cholestatic liver injury (CLI). FXR and SHP have overlapping and nonoverlapping functions in bile acid homeostasis. However, the key role played by SHP in CLI is unclear. METHODS: In this study, an alpha-naphthylisothiocyanate (ANIT)-induced cholestasis mouse model was established. The effect of SHP knockout (SHP-KO) on liver and ileal pathology was evaluated. 16S rRNA gene sequencing analysis combined with untargeted metabolomics was applied to reveal the involvement of SHP in the pathogenesis of CLI. RESULTS: The results showed that ANIT (75 mg/kg) induced cholestasis in WT mice. No significant morphological changes were found in the liver and ileal tissue of SHP-KO mice. However, the serum metabolism and intestinal flora characteristics were significantly changed. Moreover, compared with the WT + ANIT group, the serum levels of ALT and AST in the SHP-KO + ANIT group were significantly increased, and punctate necrosis in the liver tissue was more obvious. The ileum villi showed obvious shedding, thinning, and shortening. In addition, SHP-KO-associated differential intestinal flora and differential biomarkers were significantly associated. CONCLUSION: In this study, we elucidated the serum metabolic characteristics and intestinal flora changes related to the aggravation of CLI in SHP-KO mice induced by ANIT.


Subject(s)
1-Naphthylisothiocyanate , Cholestasis , Disease Models, Animal , Disease Progression , Liver , Mice, Knockout , Receptors, Cytoplasmic and Nuclear , Animals , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Cholestasis/metabolism , Cholestasis/pathology , Liver/pathology , Liver/metabolism , 1-Naphthylisothiocyanate/toxicity , Male , Ileum/pathology , Ileum/metabolism , Gastrointestinal Microbiome , Mice , Bile Acids and Salts/metabolism , Mice, Inbred C57BL
3.
Adv Exp Med Biol ; 1090: 145-163, 2018.
Article in English | MEDLINE | ID: mdl-30390289

ABSTRACT

Leptin is an adipocyte-derived hormone, which contributes to the homeostatic regulation of energy balance and metabolism through humoral and neural pathways. Leptin acts on the neurons in certain brain areas such as the hypothalamus, hippocampus, and brain stem to regulate food intake, thermogenesis, energy expenditure, and homeostasis of glucose/lipid metabolism. The pathologically increased circulating leptin is a biomarker of leptin resistance, which is common in obese individuals. Leptin resistance is defined by a reduced sensitivity or a failure in response of the brain to leptin, showing a decrease in the ability of leptin to suppress appetite or enhance energy expenditure, which causes an increased food intake and finally leads to overweight, obesity, cardiovascular diseases, and other metabolic disorders. Leptin resistance is a challenge for clinical treatment or drug discovery of obesity. Until recently, emerging evidence has been showing novel mechanisms of the leptin resistance. Here, we summarized the advances and controversy of leptin resistance and associated diseases, for better understanding the physiology and pathophysiology of leptin as well as the new strategies for treating obesity and metabolic disorders.


Subject(s)
Energy Metabolism , Homeostasis , Leptin/physiology , Brain/physiology , Humans , Obesity/pathology
4.
Adv Exp Med Biol ; 1090: 123-144, 2018.
Article in English | MEDLINE | ID: mdl-30390288

ABSTRACT

Leptin plays a critical role in the regulation of energy balance and metabolic homeostasis. Impairment of leptin signaling is closely involved in the pathogenesis of obesity and metabolic diseases, including diabetes, cardiovascular disease, etc. Leptin initiates its intracellular signaling in the leptin-receptor-expressing neurons in the central nervous system to exert physiological function, thereby leading to a suppression of appetite, a reduction of food intake, a promotion of mitochondrial oxidation, an enhancement of thermogenesis, and a decrease in body weight. In this review, the studies on leptin neural and cellular pathways are summarized with an emphasis on the progress made during the last 10 years, for better understanding the molecular mechanism of obesity and other metabolic diseases.


Subject(s)
Leptin/physiology , Receptors, Leptin/physiology , Signal Transduction , Energy Metabolism , Homeostasis , Humans , Obesity/pathology
5.
Proc Natl Acad Sci U S A ; 110(36): 14765-70, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23964123

ABSTRACT

Diphtheria toxin-mediated, acute ablation of hypothalamic neurons expressing agouti-related protein (AgRP) in adult mice leads to anorexia and starvation within 7 d that is caused by hyperactivity of neurons within the parabrachial nucleus (PBN). Because NMDA glutamate receptors are involved in various synaptic plasticity-based behavioral modifications, we hypothesized that modulation of the NR2A and NR2B subunits of the NMDA receptor in PBN neurons could contribute to the anorexia phenotype. We observed by Western blot analyses that ablation of AgRP neurons results in enhanced expression of NR2B along with a modest suppression of NR2A. Interestingly, systemic administration of LiCl in a critical time window before AgRP neuron ablation abolished the anorectic response. LiCl treatment suppressed NR2B levels in the PBN and ameliorated the local Fos induction that is associated with anorexia. This protective role of LiCl on feeding was blunted in vagotomized mice. Chronic infusion of RO25-6981, a selective NR2B inhibitor, into the PBN recapitulated the role of LiCl in maintaining feeding after AgRP neuron ablation. We suggest that the accumulation of NR2B subunits in the PBN contributes to aphagia in response to AgRP neuron ablation and may be involved in other forms of anorexia.


Subject(s)
Appetite/physiology , Neurons/physiology , Pons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Adjuvants, Immunologic/pharmacology , Agouti-Related Protein/deficiency , Agouti-Related Protein/genetics , Animals , Anorexia/genetics , Anorexia/physiopathology , Anorexia/prevention & control , Appetite/drug effects , Blotting, Western , Body Weight/drug effects , Body Weight/physiology , Deglutition Disorders/genetics , Deglutition Disorders/physiopathology , Deglutition Disorders/prevention & control , Eating/drug effects , Eating/physiology , Lithium Chloride/pharmacology , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Phenols , Piperidines/pharmacology , Pons/cytology , Pons/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Rhombencephalon/cytology , Rhombencephalon/metabolism , Rhombencephalon/physiology , Time Factors , Vagotomy
6.
Proc Natl Acad Sci U S A ; 110(17): E1631-40, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23569242

ABSTRACT

Targeted disruption of RIIß-protein kinase A (PKA) in mice leads to a lean phenotype, increased nocturnal locomotor activity, and activation of brown adipose tissue. Because RIIß is abundantly expressed in both white and brown adipose tissue as well as the brain, the contribution of neuronal vs. peripheral PKA to these phenotypes was investigated. We used a Cre-Lox strategy to reexpress RIIß in a tissue-specific manner in either adipocytes or neurons. Mice with adipocyte-specific RIIß reexpression remained hyperactive and lean, but pan-neuronal RIIß reexpression reversed both phenotypes. Selective RIIß reexpression in all striatal medium spiny neurons with Darpp32-Cre corrected the hyperlocomotor phenotype, but the mice remained lean. Further analysis revealed that RIIß reexpression in D2 dopamine receptor-expressing medium spiny neurons corrected the hyperlocomotor phenotype, which demonstrated that the lean phenotype in RIIß-PKA-deficient mice does not develop because of increased locomotor activity. To identify the neurons responsible for the lean phenotype, we used specific Cre-driver mice to reexpress RIIß in agouti-related peptide (AgRP)-, proopiomelanocortin (POMC)-, single-minded 1 (Sim1)-, or steroidogenic factor 1 (SF1)-expressing neurons in the hypothalamus, but observed no rescue of the lean phenotype. However, when RIIß was reexpressed in multiple regions of the hypothalamus and striatum driven by Rip2-Cre, or specifically in GABAergic neurons driven by Vgat-ires-Cre, both the hyperactive and lean phenotypes were completely corrected. Bilateral injection of adeno-associated virus1 (AAV1)-Cre directly into the hypothalamus caused reexpression of RIIß and partially reversed the lean phenotype. These data demonstrate that RIIß-PKA deficiency in a subset of hypothalamic GABAergic neurons leads to the lean phenotype.


Subject(s)
Adiposity/genetics , Brain/metabolism , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/metabolism , Energy Metabolism/physiology , Homeostasis/physiology , Locomotion/physiology , Neurons/metabolism , Analysis of Variance , Animals , Blotting, Western , Body Weight/genetics , Calorimetry, Indirect , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/genetics , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Genotype , Immunohistochemistry , Integrases/metabolism , Leptin/blood , Mice , Mice, Knockout , Neurons/physiology , Polymerase Chain Reaction , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
7.
Sheng Li Ke Xue Jin Zhan ; 47(4): 241-8, 2016 Aug.
Article in Zh | MEDLINE | ID: mdl-29888893

ABSTRACT

Striatum is the central structure controlling movement. It plays a pivotal role in the regulation of voluntary movement, unconscious movement, muscle tone, posture adjustment and fine movement. Dysfunction of striatum causes a variety of movement disorders ranging from the hypokinetic disorders with increased muscle tone, such as Parkinson's disease, to the hyperkinetic disorders with decreased muscle tone, such as Huntington's disease. It is generally recognized that striatum receives the neural movement signals from the motor cortex, and then processes and modifies these signals and subsequently transfers the signals back to the motor cortex via thalamus for execution of the movement through pyramidal system. The movement control function of striatum depends on a complex neural circuit system. In this review, the studies on the movement control function of striatum as well as the striatal neural circuit system are summarized with an emphasis on the progress made during recent years for better understanding the mechanism underlying the movement control function as well as the disease association of striatum.


Subject(s)
Corpus Striatum , Neural Pathways , Parkinson Disease/physiopathology , Basal Ganglia , Humans
8.
Sheng Li Ke Xue Jin Zhan ; 47(5): 321-9, 2016 10.
Article in Zh | MEDLINE | ID: mdl-29893530

ABSTRACT

Protein kinase A(PKA),as a pivotal factor in the cellular signal transduction,plays an es-sential role in the regulation of lipid metabolism.PKA activates the key lipases including hormone sensi-tive lipase (HSL)and adipose triglyceride lipase (ATGL)to promote the fat mobilization.PKA signaling up-regulates the mitochondrial thermogenesis by enhancing the expression of uncoupling protein-1 (UCP-1),which critically contributes to the body heat production.PKA is closely involved in the regulation of lipogenesis in the liver.Notably,the dysregulation of PKA signaling is associated with the pathogenic mechanisms underlying the obesity,cardiovascular diseases and diabetes mellitus.The pharmacological studies show that PKA is linked to the pharmacological effects of the major lipid regulating agents.In this review,the studies on roles of PKA in the regulation of lipid metabolism are summarized with an emphasis on progress made during the last five years for providing insights into the mechanism by which PKA regu-lates the lipid metabolism as well as the novel therapeutic strategy for lipid-metabolic diseases.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Lipid Metabolism , Uncoupling Protein 1/physiology , Animals , Diabetes Mellitus , Diagnostic Techniques, Cardiovascular , Lipase/metabolism , Lipogenesis , Obesity , Signal Transduction
9.
J Neurosci ; 34(14): 4896-904, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24695708

ABSTRACT

Striatal medium spiny neurons (MSNs) mediate many of the physiological effects of dopamine, including the regulation of feeding and motor behaviors. Dopaminergic inputs from the midbrain modulate MSN excitability through pathways that involve cAMP and protein kinase A (PKA), but the physiological role of specific PKA isoforms in MSN neurons remains poorly understood. One of the major PKA regulatory (R) subunit isoforms expressed in MSNs is RIIß, which localizes the PKA holoenzyme primarily to dendrites by interaction with AKAP5 and other scaffolding proteins. However, RI (RIα and RIß) subunits are also expressed in MSNs and the RI holoenzyme has a weaker affinity for most scaffolding proteins and tends to localize in the cell body. We generated mice with selective expression of a dominant-negative RI subunit (RIαB) in striatal MSNs and show that this dominant-negative RIαB localizes to the cytoplasm and specifically inhibits type I PKA activity in the striatum. These mice are normal at birth; however, soon after weaning they exhibit growth retardation and the adult mice are hypophagic, lean, and resistant to high-fat diet-induced hyperphagia and obesity. The RIαB-expressing mice also exhibit decreased locomotor activity and decreased dopamine-regulated CREB phosphorylation and c-fos gene expression in the striatum. Our results demonstrate a critical role for cytoplasmic RI-PKA holoenzyme in gene regulation and the overall physiological function of MSNs.


Subject(s)
Corpus Striatum/cytology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Feeding Behavior/physiology , Gene Expression Regulation/physiology , Motor Activity/physiology , Neurons/metabolism , Animals , Animals, Newborn , Body Weight/genetics , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Dopamine Agents/pharmacology , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Movement Disorders/genetics , Mutation/genetics , Obesity/diet therapy , Obesity/genetics
10.
Sheng Li Ke Xue Jin Zhan ; 46(5): 327-33, 2015 Oct.
Article in Zh | MEDLINE | ID: mdl-26904854

ABSTRACT

Leptin plays a critical role in the regulation of energy balance and metabolic homeostasis. Impairment of leptin function is closely involved in the pathogenesis of obesity, diabetes mellitus and some other metabolic diseases. Leptin initiates intracellular signal transductions in the leptin-receptor-expressing neurons in the central nervous system to exert its physiological functions. The fact that high circulating levels of leptin partially or completely fail to promote weight loss in obesity has given rise to the notion of "leptin resistance". Recently, the impairment of leptin signalings in the hypothalamus has been regarded as a critical contributor to leptin resistance. In this review, the studies on leptin signaling and leptin resistance are summarized with an emphasis on the progress made during the last five years.


Subject(s)
Signal Transduction , Central Nervous System , Energy Metabolism , Gene Expression , Homeostasis , Hypothalamus , Leptin , Neurons , Obesity
11.
Biochem Biophys Res Commun ; 450(1): 604-9, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24931672

ABSTRACT

14,15-Epoxyeicosatrienoic acid (14,15-EET), a metabolite of arachidonic acid, is enriched in the brain cortex and exerts protective effect against neuronal apoptosis induced by ischemia/reperfusion. Although apoptosis has been well recognized to be closely associated with mitochondrial biogenesis and function, it is still unclear whether the neuroprotective effect of 14,15-EET is mediated by promotion of mitochondrial biogenesis and function in cortical neurons under the condition of oxygen-glucose deprivation (OGD). In this study, we found that 14,15-EET improved cell viability and inhibited apoptosis of cortical neurons. 14,15-EET significantly increased the mitochondrial mass and the ratio of mitochondrial DNA to nuclear DNA. Key makers of mitochondrial biogenesis, peroxisome proliferator activator receptor gamma-coactivator 1 alpha (PGC-1α), nuclear respiratory factor 1 (NRF-1) and mitochondrial transcription factor A (TFAM), were elevated at both mRNA and protein levels in the cortical neurons treated with 14,15-EET. Moreover, 14,15-EET markedly attenuated the decline of mitochondrial membrane potential, reduced ROS, while increased ATP synthesis. Knockdown of cAMP-response element binding protein (CREB) by siRNA blunted the up-regulation of PGC-1α and NRF-1 stimulated by 14,15-EET, and consequently abolished the neuroprotective effect of 14,15-EET. Our results indicate that 14,15-EET protects neurons from OGD-induced apoptosis by promoting mitochondrial biogenesis and function through CREB mediated activation of PGC-1α and NRF-1.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Cerebral Cortex/physiology , Glucose/metabolism , Mitochondria/physiology , Neurons/cytology , Neurons/metabolism , Oxygen/metabolism , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Apoptosis/physiology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Neuroprotective Agents/pharmacology
12.
Cell Rep ; 43(5): 114142, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38691458

ABSTRACT

Despite medical advances, there remains an unmet need for better treatment of obesity. Itaconate, a product of the decarboxylation of the tricarboxylic acid cycle intermediate cis-aconitate, plays a regulatory role in both metabolism and immunity. Here, we show that itaconate, as an endogenous compound, counteracts high-fat-diet (HFD)-induced obesity through leptin-independent mechanisms in three mouse models. Specifically, itaconate reduces weight gain, reverses hyperlipidemia, and improves glucose tolerance in HFD-fed mice. Additionally, itaconate enhances energy expenditure and the thermogenic capacity of brown adipose tissue (BAT). Unbiased proteomic analysis reveals that itaconate upregulates key proteins involved in fatty acid oxidation and represses the expression of lipogenic genes. Itaconate may provoke a major metabolic reprogramming by inducing fatty acid oxidation and suppression of fatty acid synthesis in BAT. These findings highlight itaconate as a potential activator of BAT-mediated thermogenesis and a promising candidate for anti-obesity therapy.


Subject(s)
Adipocytes, Brown , Diet, High-Fat , Mice, Inbred C57BL , Obesity , Succinates , Thermogenesis , Animals , Thermogenesis/drug effects , Obesity/metabolism , Obesity/drug therapy , Succinates/pharmacology , Diet, High-Fat/adverse effects , Mice , Male , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Energy Metabolism/drug effects
13.
Front Pharmacol ; 14: 1197847, 2023.
Article in English | MEDLINE | ID: mdl-37284301

ABSTRACT

Background: Farnesoid X receptor (FXR) is a key metabolic target of bile acids (BAs) and is also a target for drugs against several liver diseases. However, the contribution of FXR in the pathogenesis of cholestasis is still not fully understood. The purpose of this study is to provide a comprehensive insight into the metabolic properties of FXR-involved cholestasis in mice. Materials and methods: In this study, an alpha-naphthylisothiocyanate (ANIT)-induced cholestasis mouse model and FXR-/- mice were established to investigate the effect of FXR on cholestasis. The effect of FXR on liver and ileal pathology was evaluated. Simultaneously, Untargeted metabolomics combined with 16s rRNA gene sequencing analysis was applied to reveal the involvement of FXR in the pathogenesis of cholestasis. Results: The results showed that ANIT (75 mg/kg) induced marked cholestasis in WT and FXR -/- mice. It is noteworthy that FXR-/- mice developed spontaneous cholestasis. Compared with WT mice, significant liver and ileal tissue damage were found. In addition, 16s rRNA gene sequencing analysis revealed gut microbiota dysbiosis in FXR-/- mice and ANIT-induced cholestasis mice. Differential biomarkers associated with the pathogenesis of cholestasis caused by FXR knockout were screened using untargeted metabolomics. Notably, Lactobacillus_ johnsonii_FI9785 has a high correlation with the differential biomarkers associated with the pathogenesis and progression of cholestasis caused by FXR knockout. Conclusion: Our results implied that the disorder of the intestinal flora caused by FXR knockout can also interfere with the metabolism. This study provides novel insights into the FXR-related mechanisms of cholestasis.

14.
Biology (Basel) ; 12(6)2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37372118

ABSTRACT

Inter-tissue communication (ITC) is critical for maintaining the physiological functions of multiple tissues and is closely related to the onset and development of various complex diseases. Nevertheless, there is no well-organized data resource for known ITC molecules with explicit ITC routes from source tissues to target tissues. To address this issue, in this work, we manually reviewed nearly 190,000 publications and identified 1408 experimentally supported ITC entries in which the ITC molecules, their communication routes, and their functional annotations were included. To facilitate our work, these curated ITC entries were incorporated into a user-friendly database named IntiCom-DB. This database also enables visualization of the expression abundances of ITC proteins and their interaction partners. Finally, bioinformatics analyses on these data revealed common biological characteristics of the ITC molecules. For example, tissue specificity scores of ITC molecules at the protein level are often higher than those at the mRNA level in the target tissues. Moreover, the ITC molecules and their interaction partners are more abundant in both the source tissues and the target tissues. IntiCom-DB is freely available as an online database. As the first comprehensive database of ITC molecules with explicit ITC routes to the best of our knowledge, we hope that IntiCom-DB will benefit future ITC-related studies.

15.
Adv Sci (Weinh) ; 10(5): e2205173, 2023 02.
Article in English | MEDLINE | ID: mdl-36529950

ABSTRACT

The RIIß subunit of  cAMP-dependent protein kinase A (PKA) is expressed in the brain and adipose tissue. RIIß-knockout mice show leanness and increased UCP1 in brown adipose tissue. The authors have previously reported that RIIß reexpression in hypothalamic GABAergic neurons rescues the leanness. However, whether white adipose tissue (WAT) browning contributes to the leanness and whether RIIß-PKA in these neurons governs WAT browning are unknown. Here, this work reports that RIIß-KO mice exhibit a robust WAT browning. RIIß reexpression in dorsal median hypothalamic GABAergic neurons (DMH GABAergic neurons) abrogates WAT browning. Single-cell sequencing, transcriptome sequencing, and electrophysiological studies show increased GABAergic activity in DMH GABAergic neurons of RIIß-KO mice. Activation of DMH GABAergic neurons or inhibition of PKA in these neurons elicits WAT browning and thus lowers body weight. These findings reveal that RIIß-PKA in DMH GABAergic neurons regulates WAT browning. Targeting RIIß-PKA in DMH GABAergic neurons may offer a clinically useful way to promote WAT browning for treating obesity and other metabolic disorders.


Subject(s)
Adipose Tissue, Brown , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit , Hypothalamus , Animals , Mice , Adipose Tissue, Brown/metabolism , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , GABAergic Neurons/metabolism , Hypothalamus/metabolism , Obesity/metabolism , Thinness/metabolism
16.
Sci Adv ; 9(44): eadh2884, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37910621

ABSTRACT

Overeating disorders largely contribute to worldwide incidences of obesity. Available treatments are limited. Here, we discovered that long-term chemogenetic activation of ventrolateral periaqueductal gray (vlPAG) GABAergic cells rescue obesity of high-fat diet-induced obesity (DIO) mice. This was associated with the recovery of enhanced mIPSCs, decreased food intake, increased energy expenditure, and inguinal white adipose tissue (iWAT) browning. In vivo calcium imaging confirmed vlPAG GABAergic suppression for DIO mice, with corresponding reduction in intrinsic excitability. Single-nucleus RNA sequencing identified transcriptional expression changes in GABAergic cell subtypes in DIO mice, highlighting Cacna2d1 as of potential importance. Overexpressing CACNA2D1 in vlPAG GABAergic cells of DIO mice rescued enhanced mIPSCs and calcium response, reversed obesity, and therefore presented here as a potential target for obesity treatment.


Subject(s)
Calcium , Diet, High-Fat , Mice , Animals , Diet, High-Fat/adverse effects , Calcium/metabolism , Obesity/etiology , Obesity/metabolism , Adipose Tissue, White/metabolism , Mesencephalon , Mice, Inbred C57BL
17.
Nat Commun ; 14(1): 4924, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37582782

ABSTRACT

Thermal homeostasis is vital for mammals and is controlled by brain neurocircuits. Yet, the neural pathways responsible for cold defense regulation are still unclear. Here, we found that a pathway from the lateral parabrachial nucleus (LPB) to the dorsomedial hypothalamus (DMH), which runs parallel to the canonical LPB to preoptic area (POA) pathway, is also crucial for cold defense. Together, these pathways make an equivalent and cumulative contribution, forming a parallel circuit. Specifically, activation of the LPB → DMH pathway induced strong cold-defense responses, including increases in thermogenesis of brown adipose tissue (BAT), muscle shivering, heart rate, and locomotion. Further, we identified somatostatin neurons in the LPB that target DMH to promote BAT thermogenesis. Therefore, we reveal a parallel circuit governing cold defense in mice, which enables resilience to hypothermia and provides a scalable and robust network in heat production, reshaping our understanding of neural circuit regulation of homeostatic behaviors.


Subject(s)
Hypothermia , Thermogenesis , Mice , Animals , Thermogenesis/physiology , Preoptic Area/metabolism , Neural Pathways/physiology , Homeostasis , Hypothermia/metabolism , Adipose Tissue, Brown/metabolism , Cold Temperature , Mammals
18.
Med Rev (Berl) ; 2(4): 363-384, 2022 Aug.
Article in English | MEDLINE | ID: mdl-37724323

ABSTRACT

With the prevalence of obesity and associated comorbidities, studies aimed at revealing mechanisms that regulate energy homeostasis have gained increasing interest. In 1994, the cloning of leptin was a milestone in metabolic research. As an adipocytokine, leptin governs food intake and energy homeostasis through leptin receptors (LepR) in the brain. The failure of increased leptin levels to suppress feeding and elevate energy expenditure is referred to as leptin resistance, which encompasses complex pathophysiological processes. Within the brain, LepR-expressing neurons are distributed in hypothalamus and other brain areas, and each population of the LepR-expressing neurons may mediate particular aspects of leptin effects. In LepR-expressing neurons, the binding of leptin to LepR initiates multiple signaling cascades including janus kinase (JAK)-signal transducers and activators of transcription (STAT) phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT), extracellular regulated protein kinase (ERK), and AMP-activated protein kinase (AMPK) signaling, etc., mediating leptin actions. These findings place leptin at the intersection of metabolic and neuroendocrine regulations, and render leptin a key target for treating obesity and associated comorbidities. This review highlights the main discoveries that shaped the field of leptin for better understanding of the mechanism governing metabolic homeostasis, and guides the development of safe and effective interventions to treat obesity and associated diseases.

19.
Diabetes ; 71(2): 249-263, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34732538

ABSTRACT

The increasing prevalence of obesity has resulted in demands for the development of new effective strategies for obesity treatment. Withaferin A (WA) shows a great potential for prevention of obesity by sensitizing leptin signaling in the hypothalamus. However, the mechanism underlying the weight- and adiposity-reducing effects of WA remains to be elucidated. In this study, we report that WA treatment induced white adipose tissue (WAT) browning, elevated energy expenditure, decreased respiratory exchange ratio, and prevented high-fat diet-induced obesity. The sympathetic chemical denervation dampened the WAT browning and also impeded the reduction of adiposity in WA-treated mice. WA markedly upregulated the levels of Prdm16 and FATP1 (Slc27a1) in the inguinal WAT (iWAT), and this was blocked by sympathetic denervation. Prdm16 or FATP1 knockdown in iWAT abrogated the WAT browning-inducing effects of WA and restored the weight gain and adiposity in WA-treated mice. Together, these findings suggest that WA induces WAT browning through the sympathetic nerve-adipose axis, and the adipocytic Prdm16-FATP1 pathway mediates the promotive effects of WA on white adipose browning.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Obesity/prevention & control , Withanolides/pharmacology , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/physiology , Adipose Tissue, White/innervation , Adipose Tissue, White/physiology , Animals , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet, High-Fat , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Nutrients ; 14(17)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36079775

ABSTRACT

Anxiety is a common disorder among college students, especially those with obesity. Obesity contributes to metabolic disorders and disturbs the neural functions, further leading to anxiety. In this cross-sectional study, we aimed to determine the association between obesity and anxiety among college students and identified the potential factors for obesity-associated anxiety. We evaluated the intervention effects of calorie restriction on anxiety. Self-reported questionnaires were distributed to 1381 college students from January to March in 2021. Anxiety was measured by the State-Trait Anxiety Inventory (STAI). Participants were classified into anxiety and non-anxiety groups according to their STAI scores. Chi-squared test and logistic regression were used to analyze the potential factors. We found that 383 college students exhibited anxiety, accounting for 30.1% among all included college students, which was higher than the global average. The association between anxiety and obesity was observed among college students (p = 0.009), especially in males (p = 0.007). We identified that pre-obesity (p = 0.012), unhealthy calorie intake (p = 0.001), dieting (p = 0.003) and high academic year (p = 0.006) as the risk factors for anxiety and found that the long sleep duration was a protective factor for anxiety (p < 0.001). We found that more obese students showed an improvement of anxiety than the underweight students after calorie restriction (p < 0.001). Collectively, our findings suggest that obesity-associated anxiety is prevalent among the college students and could be alleviated by moderate calorie restriction. It is necessary for students to receive anxiety management in their college life. Additionally, the proper calorie restriction should be promoted to help students protect against obesity and obesity-associated anxiety.


Subject(s)
Caloric Restriction , Students , Anxiety/epidemiology , Cross-Sectional Studies , Humans , Male , Obesity/complications , Obesity/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL