{"title":"Intestinal butyric acid-mediated disruption of gut hormone secretion and lipid metabolism in vasopressin receptor-deficient mice.","authors":"Kazuki Harada, Eiji Wada, Yuri Osuga, Kie Shimizu, Reiko Uenoyama, Masami Yokota Hirai, Fumihiko Maekawa, Masao Miyazaki, Yukiko K Hayashi, Kazuaki Nakamura, Takashi Tsuboi","doi":"10.1016/j.molmet.2024.102072","DOIUrl":"10.1016/j.molmet.2024.102072","url":null,"abstract":"<p><strong>Objectives: </strong>Arginine vasopressin (AVP), known as an antidiuretic hormone, is also crucial in metabolic homeostasis. Although AVP receptor-deficient mice exhibit various abnormalities in glucose and lipid metabolism, the mechanism underlying these symptoms remains unclear. This study aimed to explore the involvement of the gut hormones including glucagon-like peptide-1 (GLP-1) and microbiota as essential mediators.</p><p><strong>Methods: </strong>We used the mouse GLP-1-secreting cell line, GLUTag, and performed live cell imaging to examine the contribution of V1a and V1b vasopressin receptors (V1aR and V1bR, respectively) to GLP-1 secretion. We next investigated the hormone dynamics of V1aR-deficient mice (V1aR<sup>-/-</sup> mice), V1bR-deficient mice (V1bR<sup>-/-</sup> mice), and V1aR/V1bR-double deficient mice (V1aR<sup>-/-</sup>V1bR<sup>-/-</sup>mice).</p><p><strong>Results: </strong>AVP induced the increase in intracellular Ca<sup>2+</sup> levels and GLP-1 secretion from GLUTag cells in a V1aR and V1bR-dependent manner. AVP receptor-deficient mice, particularly V1aR<sup>-/-</sup>V1bR<sup>-/-</sup> mice, demonstrated impaired secretion of GLP-1 and peptide YY secreted by enteroendocrine L cells. V1aR<sup>-/-</sup>V1bR<sup>-/-</sup>mice also exhibited abnormal lipid accumulation in the brown adipose tissue and skeletal muscle. We discovered that V1aR<sup>-/-</sup>V1bR<sup>-/-</sup> mice showed increased Paneth cell-related gene expression in the small intestine, which was attributed to increased fecal butyric acid levels. Exposure to butyric acid reduced GLP-1 secretion in L cell line. Additionally, human Paneth cell-related gene expressions negatively correlated with that of V1 receptor genes.</p><p><strong>Conclusions: </strong>The deficiency in V1 receptor genes may increase gut butyric acid levels and impair the function of L cells, thus dysregulating lipid homeostasis in the brown adipose tissue and skeletal muscle. This study highlights the importance of appropriate control of the gut microbiota and its metabolites, including butyric acid, for the optimum functioning of enteroendocrine cells.</p>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":" ","pages":"102072"},"PeriodicalIF":7.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11728074/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142818469","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular MetabolismPub Date : 2025-01-01Epub Date: 2024-11-25DOI: 10.1016/j.molmet.2024.102073
William Roell, Tamer Coskun, Teayoun Kim, Libbey O'Farrell, Jennifer A Martin, Shelly Nason, Jasmin Hernandez-Alamillo, Saidharshana Dhantu, Daniel J Drucker, Kyle W Sloop, James P Steele, Jorge Alsina-Fernandez, Kirk M Habegger
{"title":"Characterization of LY3324954 a long-acting glucagon-receptor agonist.","authors":"William Roell, Tamer Coskun, Teayoun Kim, Libbey O'Farrell, Jennifer A Martin, Shelly Nason, Jasmin Hernandez-Alamillo, Saidharshana Dhantu, Daniel J Drucker, Kyle W Sloop, James P Steele, Jorge Alsina-Fernandez, Kirk M Habegger","doi":"10.1016/j.molmet.2024.102073","DOIUrl":"10.1016/j.molmet.2024.102073","url":null,"abstract":"<p><strong>Objective: </strong>Glucagon is a crucial regulator of glucose and lipid metabolism as well as whole-body energy balance. Thus, modulation of glucagon receptor (GCGR) activity in the context of single-molecule multi-receptor co-agonists has become an emerging therapeutic target against obesity and obesity-associated metabolic dysfunction. To better elucidate the role of GCGR-signaling when paired with incretin receptor signaling or on its own, we developed, LY3324954, a GCGR agonist with improved potency and selectivity as compared to the native glucagon peptide.</p><p><strong>Methods: </strong>LY3324954 was administered to DIO mice, rats, dogs, and monkeys to evaluate pharmacokinetic (PK) profile. Biweekly treatments were conducted in lean and DIO mice to characterize LY3324954-effects on glucose homeostasis and energy balance. Single dose studies were also conducted in liver Gcgr-deficient mice to establish receptor specificity.</p><p><strong>Results: </strong>LY3324954 also exhibited extended PK profile in DIO mice, rats, dogs, and monkeys. When administered every 72 h, LY3324954 treatment stimulated transient glucose and insulin excursions in lean mice. In diet-induced obese mice, LY3324954 treatment stimulates energy expenditure, weight loss, and a reduction of adiposity in a dose-dependent manner. Benefit to whole-body lipid homeostasis was likewise observed in these mice.</p><p><strong>Conclusions: </strong>Taken together, these studies characterize a long-acting and potent GCGR-agonist and its regulation of glucose and lipid metabolism as well as whole-body energy balance following both acute and chronic treatment in mice.</p>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":" ","pages":"102073"},"PeriodicalIF":7.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11696851/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142739909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xiaomin Luo , Fang Liu , Lijun Zhu , Caizhi Liu , Ruhui Shen , Xiaoyin Ding , Yufan Wang , Xiaofang Tang , Yongde Peng , Zhijian Zhang
{"title":"Leupaxin promotes hepatic gluconeogenesis and glucose metabolism by coactivation with hepatic nuclear factor 4α","authors":"Xiaomin Luo , Fang Liu , Lijun Zhu , Caizhi Liu , Ruhui Shen , Xiaoyin Ding , Yufan Wang , Xiaofang Tang , Yongde Peng , Zhijian Zhang","doi":"10.1016/j.molmet.2024.102075","DOIUrl":"10.1016/j.molmet.2024.102075","url":null,"abstract":"<div><h3>Background</h3><div>As the primary source of glucose during fasting, hepatic gluconeogenesis is rigorously regulated to maintain euglycemia. Abnormal gluconeogenesis in the liver can lead to hyperglycemia, a key diagnostic marker and the primary pathological contributor to type 2 diabetes (T2D) and metabolic disorders. Hepatic nuclear factor-4 (HNF4α) is an important regulator of gluconeogenesis. In this study, we identify leupaxin (LPXN) as a novel coactivator for HNF4α. Although previous studies have shown that LPXN is highly correlated with cancer types such as B-cell differentiation and hepatocellular carcinoma progression, the role of LPXN in gluconeogenesis remains unknown.</div></div><div><h3>Methods</h3><div>We initially used protein pull-down assays, mass spectrometry and luciferase assays to identify the coactivator that interacts with HNF4α in gluconeogenesis. We further leveraged cell cultures and mouse models to validate the functional importance of molecular pathway during gluconeogenesis by using adenovirus-mediated overexpression and adeno-associated virus shRNA–mediated knockdown both <em>in vivo</em> and <em>ex vivo,</em> such as in ob/db/DIO mice, HepG2 and primary hepatocytes. Following, we used CUT&Tag and chip qPCR to identify the LPXN-mediated mechanisms underlying the observed abnormal gluconeogenesis. Additionally, we assessed the translational relevance of our findings using human liver tissues from both healthy donors and patients with obesity/type 2 diabetes.</div></div><div><h3>Results</h3><div>We found that LPXN interacts with HNF4α to participate in gluconeogenesis. Knockdown of LPXN expression in the liver effectively enhanced glucose metabolism, while its overexpression in the liver effectively inhibited it. Mechanistically, LPXN could translocate into the nucleus and was essential for regulating gluconeogenesis by binding to the PEPCK promoter, which controlled the expression of an enzyme involved in gluconeogenesis, mainly through the Gcg-cAMP-PKA pathway. Additionally, LPXN expression was found to be increased in the livers of patients with steatosis and diabetes, supporting a pathological role of LPXN.</div></div><div><h3>Conclusions</h3><div>Taken together, our study provides evidence that LPXN plays a critical role in modulating hepatic gluconeogenesis, thereby reinforcing the fact that targeting LPXN may be a potential approach for the treatment of diabetes and metabolic disorders.</div></div>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":"91 ","pages":"Article 102075"},"PeriodicalIF":7.0,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142739911","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cecilia Hes , Lu Ting Gui , Alexandre Bay , Fernando Alvarez , Pierce Katz , Tanushree Paul , Nadejda Bozadjieva-Kramer , Randy J. Seeley , Ciriaco A. Piccirillo , Paul V. Sabatini
{"title":"GDNF family receptor alpha-like (GFRAL) expression is restricted to the caudal brainstem","authors":"Cecilia Hes , Lu Ting Gui , Alexandre Bay , Fernando Alvarez , Pierce Katz , Tanushree Paul , Nadejda Bozadjieva-Kramer , Randy J. Seeley , Ciriaco A. Piccirillo , Paul V. Sabatini","doi":"10.1016/j.molmet.2024.102070","DOIUrl":"10.1016/j.molmet.2024.102070","url":null,"abstract":"<div><h3>Objective</h3><div>Growth differentiation factor 15 (GDF15) acts on the receptor dimer of GDNF family receptor alpha-like (GFRAL) and Rearranged during transfection (RET). While <em>Gfral</em>-expressing cells are known to be present in the area postrema and nucleus of the solitary tract (AP/NTS) located in the brainstem, the presence of Gfral-expressing cells in other sites within the central nervous system and peripheral tissues is not been fully addressed. Our objective was to thoroughly investigate whether GFRAL is expressed in peripheral tissues and in brain sites different from the brainstem.</div></div><div><h3>Methods</h3><div>From <em>Gfral</em>:eGFP mice we collected tissue from 12 different tissues, including brain, and used single molecule <em>in-situ</em> hybridizations to identify cells within those tissues expressing <em>Gfral</em>. We then contrasted the results with human <em>Gfral</em>-expression by analyzing publicly available single-cell RNA sequencing data.</div></div><div><h3>Results</h3><div>In mice we found readably detectable <em>Gfral</em> mRNA within the AP/NTS but not within other brain sites. Within peripheral tissues, we failed to detect any <em>Gfral</em>-labelled cells in the vast majority of examined tissues and when present, were extremely rare. Single cell sequencing of human tissues confirmed <em>GFRAL</em>-expressing cells are detectable in some sites outside the AP/NTS in an extremely sparse manner. Importantly, across the utilized methodologies, smFISH, genetic <em>Gfral</em> reporter mice and scRNA-Seq, we failed to detect <em>Gfral</em>-labelled cells with all three.</div></div><div><h3>Conclusions</h3><div>Through highly sensitive and selective technologies we show <em>Gfral</em> expression is overwhelmingly restricted to the brainstem and expect that GDF15 and GFRAL-based therapies in development for cancer cachexia will specifically target AP/NTS cells.</div></div>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":"91 ","pages":"Article 102070"},"PeriodicalIF":7.0,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142750866","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Philippe Zizzari , Ashley Castellanos-Jankiewicz , Selma Yagoub , Vincent Simon , Samantha Clark , Marlene Maître , Nathalie Dupuy , Thierry Leste-Lasserre , Delphine Gonzales , Kristina Schoonjans , Valérie S. Fénelon , Daniela Cota
{"title":"TGR5 receptors in SF1-expressing neurons of the ventromedial hypothalamus regulate glucose homeostasis","authors":"Philippe Zizzari , Ashley Castellanos-Jankiewicz , Selma Yagoub , Vincent Simon , Samantha Clark , Marlene Maître , Nathalie Dupuy , Thierry Leste-Lasserre , Delphine Gonzales , Kristina Schoonjans , Valérie S. Fénelon , Daniela Cota","doi":"10.1016/j.molmet.2024.102071","DOIUrl":"10.1016/j.molmet.2024.102071","url":null,"abstract":"<div><h3>Objective</h3><div>Steroidogenic factor-1 (SF1) neurons of the ventromedial hypothalamus play key roles in the regulation of food intake, body weight and glucose metabolism. The bile acid receptor Takeda G protein-coupled receptor 5 (TGR5) is expressed in the hypothalamus, where it determines some of the actions of bile acids on food intake and body weight through still poorly defined neuronal mechanisms. Here, we examined the role of TGR5 in SF1 neurons in the regulation of energy balance and glucose metabolism.</div></div><div><h3>Methods</h3><div>We used a genetic approach combined with metabolic phenotyping and molecular analyses to establish the effect of TGR5 deletion in SF1 neurons on meal pattern, body weight, body composition, energy expenditure and use of energy substrates as well as on possible changes in glucose handling and insulin sensitivity.</div></div><div><h3>Results</h3><div>Our findings reveal that TGR5 in SF1 neurons does not play a major role in the regulation of food intake or body weight under standard chow, but it is involved in the adaptive feeding response to the acute exposure to cold or to a hypercaloric, high-fat diet, without changes in energy expenditure. Notably, TGR5 in SF1 neurons hinder glucose metabolism, since deletion of the receptor improves whole-body glucose uptake through heightened insulin signaling in the hypothalamus and in the brown adipose tissue.</div></div><div><h3>Conclusions</h3><div>TGR5 in SF1 neurons favours satiety by differently modifying the meal pattern in response to specific metabolic cues. These studies also reveal a novel key function for TGR5 in SF1 neurons in the regulation of whole-body insulin sensitivity, providing new insight into the role played by neuronal TGR5 in the regulation of metabolism.</div></div>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":"91 ","pages":"Article 102071"},"PeriodicalIF":7.0,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142738778","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jordan Wean, Allison Ho Kowalsky, Rhianna Laker, Sarah Will, Daniel J Drucker, Christopher J Rhodes, Randy J Seeley
{"title":"Specific loss of GIPR signaling in GABAergic neurons enhances GLP-1R agonist-induced body weight loss.","authors":"Jordan Wean, Allison Ho Kowalsky, Rhianna Laker, Sarah Will, Daniel J Drucker, Christopher J Rhodes, Randy J Seeley","doi":"10.1016/j.molmet.2024.102074","DOIUrl":"10.1016/j.molmet.2024.102074","url":null,"abstract":"<p><strong>Objectives: </strong>Dual incretin agonists are among the most effective pharmaceutical treatments for obesity and type 2 diabetes to date. Such therapeutics can target two receptors, such as the glucagon-like peptide-1 (GLP-1) receptor and the glucose-dependent insulinotropic polypeptide (GIP) receptor in the case of tirzepatide, to improve glycemia and reduce body weight. Regarding body weight effects, GIPR signaling is thought to involve at least two relevant mechanisms: the enhancement of food intake reduction and the attenuation of aversive effects caused by GLP-1R agonists. Although it is known that dual GLP-1R-GIPR agonism produces greater weight loss than GLP-1R agonism alone, the precise mechanism is unknown.</p><p><strong>Methods: </strong>To address this question, we used mice lacking GIPR in the whole body, GABAergic neurons, or glutamatergic neurons. These mice were given various combinations of GLP-1R and GIPR agonist drugs with subsequent food intake and conditioned taste aversion measurements.</p><p><strong>Results: </strong>A GIPR knockout in either the whole body or selectively in inhibitory GABAergic neurons protects against diet-induced obesity, whereas a knockout in excitatory glutamatergic neurons had a negligible effect. Furthermore, we found that GIPR in GABAergic neurons is essential for the enhanced weight loss efficacy of dual incretin agonism, yet, surprisingly, its removal enhances the effect of GLP-1R agonism alone. Finally, GIPR knockout in GABAergic neurons prevents the anti-aversive effects of GIPR agonism.</p><p><strong>Conclusions: </strong>Our findings are consistent with GIPR research at large in that both enhancement and removal of GIPR signaling are metabolically beneficial. Notably, however, our findings suggest that future obesity therapies designed to modulate GIPR signaling, whether by agonism or antagonism, would be best targeted towards GABAergic neurons.</p>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":" ","pages":"102074"},"PeriodicalIF":7.0,"publicationDate":"2024-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142755419","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fangzhen Xia , Ziteng Zhang , Zhen Qian , Xiaoyu Fang , Junxue Wang , Yan Wang , Guoting Sun , Yuefeng Yu , Ninjian Wang , Junke Zhen , Yan Liu , Yingli Lu
{"title":"The immune checkpoint molecule B7-H4 regulates β-cell mass and insulin secretion by modulating cholesterol metabolism through Stat5 signalling","authors":"Fangzhen Xia , Ziteng Zhang , Zhen Qian , Xiaoyu Fang , Junxue Wang , Yan Wang , Guoting Sun , Yuefeng Yu , Ninjian Wang , Junke Zhen , Yan Liu , Yingli Lu","doi":"10.1016/j.molmet.2024.102069","DOIUrl":"10.1016/j.molmet.2024.102069","url":null,"abstract":"<div><h3>Objective</h3><div>B7-H4 (B7S1, B7x, VTCN1) is an important immune checkpoint molecule that maintains immune homeostasis and is also expressed in pancreatic β cells. The polymorphism of B7-H4 influences the prevalence of Type 2 diabetes (T2D), suggesting a potential role of B7-H4 in the physiological function of pancreatic β cells and the pathogenesis of T2D.</div></div><div><h3>Methods</h3><div>β-cell-specific B7-H4 knockout mice (B7-H4 cKO mice) and their wild-type littermates were used to investigate the <em>in vivo</em> effects of B7-H4 on pancreatic β-cell morphology and function. AAV2/8-ins2-B7H4 and a control virus were infused via the pancreatic intraduct into high-fat diet (HFD)-treated mice to elucidate the therapeutic effect of B7-H4. RNA sequencing was conducted on primary islets. A Luminex assay was used to quantify cytokine changes in B7-H4 cKO mice. Electron microscopy imaging was used to observe insulin secretory vesicles in pancreatic β cells.</div></div><div><h3>Results</h3><div>Lesion of B7-H4 in β cells results in glucose intolerance due to reduced β-cell mass and deficient insulin secretion, whereas overexpression of B7-H4 in β cells ameliorates glucose intolerance in HFD-fed mice. Mechanistically, B7-H4 deficiency activates signal transducer and activator of transcription 5 (Stat5) signalling, which inhibits the expression of apolipoprotein F (Apof), leading to reduced cholesterol efflux and accumulated cholesterol in β cells, thereby impairing insulin processing and secretion. Overexpression of Apof in β cells or intraperitoneal injection of a Stat5 inhibitor reverses the metabolic phenotype and insulin secretion deficiency in B7-H4 cKO mice.</div></div><div><h3>Conclusion</h3><div>Our study demonstrated that B7-H4 plays an important role in regulating β-cell mass and insulin secretion, which may shed new light on the development of novel strategies for T2D treatment.</div></div>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":"91 ","pages":"Article 102069"},"PeriodicalIF":7.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687731","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sean Y. Ng , Andrew I. Mikhail , Stephanie R. Mattina , Salah A. Mohammed , Shahzeb K. Khan , Eric M. Desjardins , Changhyun Lim , Stuart M. Phillips , Gregory R. Steinberg , Vladimir Ljubicic
{"title":"AMPK regulates the maintenance and remodelling of the neuromuscular junction","authors":"Sean Y. Ng , Andrew I. Mikhail , Stephanie R. Mattina , Salah A. Mohammed , Shahzeb K. Khan , Eric M. Desjardins , Changhyun Lim , Stuart M. Phillips , Gregory R. Steinberg , Vladimir Ljubicic","doi":"10.1016/j.molmet.2024.102066","DOIUrl":"10.1016/j.molmet.2024.102066","url":null,"abstract":"<div><h3>Objective</h3><div>The molecular mechanisms underlying the maintenance and adaptability of the neuromuscular junction (NMJ) remain poorly understood. This study aimed to investigate the role of AMP-activated protein kinase (AMPK) as a key regulator of NMJ stability and plasticity.</div></div><div><h3>Method</h3><div>A comprehensive, multifaceted approach was employed, integrating genetic, physiological, and pharmacological methodologies to elucidate the role of skeletal muscle AMPK in modulating the neuromuscular synapse.</div></div><div><h3>Results</h3><div>Our findings reveal an increased abundance of AMPK transcripts within the NMJ and an age-associated decline in AMPK activity and synapse-specific mitochondrial gene expression. Young mice null for skeletal muscle AMPK displayed a neuromuscular phenotype akin to aged animals. Pharmacological AMPK stimulation facilitated its localization in subsynaptic myonuclei, preceded the induction of several NMJ-related transcripts, and enhanced myotube acetylcholine receptor clustering. Exercise-induced AMPK activation in mouse muscle elicited a broad NMJ-related gene response, consistent with human exercise data.</div></div><div><h3>Conclusions</h3><div>These findings highlight a critical role for AMPK in the maintenance and remodeling of the NMJ, highlighting its potential as a therapeutic target for age-related and neuromuscular disorders.</div></div>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":"91 ","pages":"Article 102066"},"PeriodicalIF":7.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687727","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Md Shahjalal H. Khan , Sora Q. Kim , Robert C. Ross , Florina Corpodean , Redin A. Spann , Diana A. Albarado , Sun O. Fernandez-Kim , Blaise Clarke , Hans-Rudolf Berthoud , Heike Münzberg , David H. McDougal , Yanlin He , Sangho Yu , Vance L. Albaugh , Paul L. Soto , Christopher D. Morrison
{"title":"FGF21 acts in the brain to drive macronutrient-specific changes in behavioral motivation and brain reward signaling","authors":"Md Shahjalal H. Khan , Sora Q. Kim , Robert C. Ross , Florina Corpodean , Redin A. Spann , Diana A. Albarado , Sun O. Fernandez-Kim , Blaise Clarke , Hans-Rudolf Berthoud , Heike Münzberg , David H. McDougal , Yanlin He , Sangho Yu , Vance L. Albaugh , Paul L. Soto , Christopher D. Morrison","doi":"10.1016/j.molmet.2024.102068","DOIUrl":"10.1016/j.molmet.2024.102068","url":null,"abstract":"<div><h3>Objective</h3><div>Dietary protein restriction induces adaptive changes in food preference, increasing protein consumption over carbohydrates or fat. We investigated whether motivation and reward signaling underpin these preferences.</div></div><div><h3>Methods and Results</h3><div>In an operant task, protein-restricted male mice responded more for liquid protein rewards, but not carbohydrate, fat, or sweet rewards compared to non-restricted mice. When the number of responses required to access protein reward varied, protein-restricted mice exhibited higher operant responses at moderate to high response requirements. The protein restriction-induced increase in operant responding for protein was absent in <em>Fgf21</em>-KO mice and mice with neuron-specific deletion of the FGF21 co-receptor beta-Klotho (Klb<sup>Cam2ka</sup>). Fiber photometry recording of VTA dopamine neurons revealed that oral delivery of maltodextrin triggered a larger dopamine neuron activation than casein in control diet-fed mice, while casein triggered a larger activation in low-protein diet-fed mice. This restriction-induced shift in nutrient-specific VTA dopamine signaling was lost in <em>Fgf21</em>-KO mice.</div></div><div><h3>Conclusion</h3><div>These data suggest that the increased FGF21 during protein restriction acts in the brain to induce a protein-specific appetite by specifically enhancing the reward value of protein-containing foods and the motivation to consume them.</div></div>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":"91 ","pages":"Article 102068"},"PeriodicalIF":7.0,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687729","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Tábatha de Oliveira Silva , Guilherme Lunardon , Caroline A. Lino , Amanda de Almeida Silva , Shiju Zhang , Maria Cláudia Costa Irigoyen , Yao Wei Lu , John D. Mably , Maria Luiza M. Barreto-Chaves , Da-Zhi Wang , Gabriela P. Diniz
{"title":"Senescent cell depletion alleviates obesity-related metabolic and cardiac disorders","authors":"Tábatha de Oliveira Silva , Guilherme Lunardon , Caroline A. Lino , Amanda de Almeida Silva , Shiju Zhang , Maria Cláudia Costa Irigoyen , Yao Wei Lu , John D. Mably , Maria Luiza M. Barreto-Chaves , Da-Zhi Wang , Gabriela P. Diniz","doi":"10.1016/j.molmet.2024.102065","DOIUrl":"10.1016/j.molmet.2024.102065","url":null,"abstract":"<div><div>Obesity is a major contributor to metabolic and cardiovascular disease. Although senescent cells have been shown to accumulate in adipose tissue, the role of senescence in obesity-induced metabolic disorders and in cardiac dysfunction is not yet clear; therefore, the therapeutic potential of managing senescence in obesity-related metabolic and cardiac disorders remains to be fully defined.</div></div><div><h3>Objective</h3><div>We investigated the beneficial effects of a senolytic cocktail (dasatinib and quercetin) on senescence and its influence on obesity-related parameters.</div></div><div><h3>Methods and Results</h3><div>We found that the increase in body weight and adiposity, glucose intolerance, insulin resistance, dyslipidemia, hyperleptinemia, and hepatic disorders which were induced by an obesogenic diet were alleviated by senolytic cocktail treatment in mice. Treatment with senolytic compounds eliminated senescent cells, counteracting the activation of the senescence program and DNA damage in white adipose tissue (WAT) observed with an obesogenic diet. Moreover, the senolytic cocktail prevented the brown adipose tissue (BAT) whitening and increased the expression of the thermogenic gene profile in BAT and pWAT. In the hearts of obese mice, senolytic combination abolished myocardial maladaptation, reducing the senescence-associated secretory phenotype (SASP) and DNA damage, repressing cardiac hypertrophy, and improving diastolic dysfunction. Additionally, we showed that treatment with the senolytic cocktail corrected gene expression programs associated with fatty acid metabolism, oxidative phosphorylation, the P53 pathway, and DNA repair, which were all downregulated in obese mice.</div></div><div><h3>Conclusions</h3><div>Collectively, these data suggest that a senolytic cocktail can prevent the activation of the senescence program in the heart and WAT and activate the thermogenic program in BAT. Our results suggest that targeting senescent cells may be a novel therapeutic strategy for alleviating obesity-related metabolic and cardiac disorders.</div></div>","PeriodicalId":18765,"journal":{"name":"Molecular Metabolism","volume":"91 ","pages":"Article 102065"},"PeriodicalIF":7.0,"publicationDate":"2024-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142667981","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}