1
|
Suryaningtyas IT, Jung WK, Lee SJ, Je JY. Bioactive peptides PIISVYWK and FSVVPSPK improve glucose homeostasis by targeting DPP-IV and glucose transport in type 2 diabetic mice. Int Immunopharmacol 2025; 158:114844. [PMID: 40359889 DOI: 10.1016/j.intimp.2025.114844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
Type 2 Diabetes Mellitus (T2DM) is a metabolic disorder characterized by chronic hyperglycemia, insulin resistance, and progressive β-cell dysfunction, often exacerbated by inflammation and oxidative stress. Effective management requires multi-targeted approaches, including modulation of glucose metabolism, suppression of inflammatory pathways, and pancreatic protection. This study investigates the antidiabetic and immunomodulatory potential of PIISVYWK (P1) and FSVVPSPK (P2), bioactive peptides from blue mussel, Mytilus edulis, in regulating these pathways. In vitro, P1 and P2 were assessed for their ability to inhibit α-glucosidase and DPP-IV activity in Caco-2 cells, alongside glucose uptake and transporter protein expression (SGLT-1 and GLUT2). In vivo, HFD/STZ-induced diabetic mice were administered P1 or P2 (1 mg/kg or 10 mg/kg) or metformin (200 mg/kg) for four weeks. Peptide treatment significantly improved glycemic control by inhibiting α-glucosidase and DPP-IV, increasing GLP-1 levels, and reducing intestinal glucose uptake. Additionally, P1 and P2 exhibited strong anti-inflammatory effects by suppressing NF-κB activation and reducing circulating IL-6, TNF-α, and IL-1β levels. Enhanced antioxidant enzyme activity (SOD, GPx, CAT) further mitigated oxidative stress, preventing pancreatic damage. Peptides also preserved β-cell function by enhancing insulin secretion and regulating glucagon levels. These findings suggest that P1 and P2 peptides exert antidiabetic effects through multi-targeted mechanisms, including immunomodulation, making them promising therapeutic candidates for T2DM management.
Collapse
Affiliation(s)
- Indyaswan T Suryaningtyas
- Research Center for Marine-Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Food Technology and Processing, National Research and Innovation Agency, Yogyakarta 55861, Indonesia
| | - Won-Kyo Jung
- Research Center for Marine-Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Sei-Jung Lee
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
2
|
AL-Noshokaty TM, Abdelhamid R, Abdelmaksoud NM, Khaled A, Hossam M, Ahmed R, Saber T, Khaled S, Elshaer SS, Abulsoud AI. Unlocking the multifaceted roles of GLP-1: Physiological functions and therapeutic potential. Toxicol Rep 2025; 14:101895. [PMID: 39911322 PMCID: PMC11795145 DOI: 10.1016/j.toxrep.2025.101895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025] Open
Abstract
Glucagon (GCG) like peptide 1 (GLP-1) has emerged as a powerful player in regulating metabolism and a promising therapeutic target for various chronic diseases. This review delves into the physiological roles of GLP-1, exploring its impact on glucose homeostasis, insulin secretion, and satiety. We examine the compelling evidence supporting GLP-1 receptor agonists (GLP-1RAs) in managing type 2 diabetes (T2D), obesity, and other diseases. The intricate molecular mechanisms underlying GLP-1RAs are explored, including their interactions with pathways like extracellular signal-regulated kinase 1/2 (ERK1/2), activated protein kinase (AMPK), cyclic adenine monophosphate (cAMP), mitogen-activated protein kinase (MAPK), and protein kinase C (PKC). Expanding our understanding, the review investigates the potential role of GLP-1 in cancers. Also, microribonucleic acid (RNA) (miRNAs), critical regulators of gene expression, are introduced as potential modulators of GLP-1 signaling. We delve into the link between miRNAs and T2D obesity and explore specific miRNA examples influencing GLP-1R function. Finally, the review explores the rationale for seeking alternatives to GLP-1RAs and highlights natural products with promising GLP-1 modulatory effects.
Collapse
Affiliation(s)
- Tohada M. AL-Noshokaty
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Rehab Abdelhamid
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | | | - Aya Khaled
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mariam Hossam
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Razan Ahmed
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Toka Saber
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Shahd Khaled
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Shereen Saeid Elshaer
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Ahmed I. Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt
- Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| |
Collapse
|
3
|
Moiz A, Filion KB, Tsoukas MA, Yu OH, Peters TM, Eisenberg MJ. Mechanisms of GLP-1 Receptor Agonist-Induced Weight Loss: A Review of Central and Peripheral Pathways in Appetite and Energy Regulation. Am J Med 2025; 138:934-940. [PMID: 39892489 DOI: 10.1016/j.amjmed.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) have become central in managing obesity and type 2 diabetes, primarily through appetite suppression and metabolic regulation. This review explores the mechanisms underlying GLP-1 RA-induced weight loss, focusing on central and peripheral pathways. Centrally, GLP-1 RAs modulate brain regions controlling appetite, influencing neurotransmitter and peptide release to regulate hunger and energy expenditure. Peripherally, GLP-1 RAs improve glycemic control by enhancing insulin secretion, reducing glucagon release, delaying gastric emptying, and regulating gut hormones. They also reduce triglycerides and low-density lipoprotein cholesterol, mitigate adipose tissue inflammation, and minimize ectopic fat deposition, promoting overall metabolic health. Emerging dual and triple co-agonists, targeting GLP-1 alongside glucose-dependent insulinotropic polypeptide, and glucagon pathways, may enhance weight loss and metabolic flexibility. Understanding these mechanisms is crucial as the therapeutic landscape evolves, offering clinicians and researchers insights to optimize the efficacy of current and future obesity treatments.
Collapse
Affiliation(s)
- Areesha Moiz
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Kristian B Filion
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada
| | - Michael A Tsoukas
- Department of Medicine, McGill University, Montreal, Canada; Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| | - Oriana Hy Yu
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| | - Tricia M Peters
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Division of Experimental Medicine, McGill University, Montreal, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| | - Mark J Eisenberg
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Division of Experimental Medicine, McGill University, Montreal, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Division of Cardiology, Jewish General Hospital/McGill University, Canada.
| |
Collapse
|
4
|
Han X, Feng T, Yang Y, Zhu Z, Shao F, Sun L, Yin Y, Zhang W. Intestinal Epithelial-Derived Exosomes Under Cold Stimulation Promote Adipose Thermogenesis. Metabolites 2025; 15:324. [PMID: 40422900 DOI: 10.3390/metabo15050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
Background: Whether intestinal epithelial cells can regulate distant adipose tissue remains a mystery. Methods: Cold-stimulated intestinal epithelial cell-derived exosomes (Cold IEC-Exo) play a pivotal role in enhancing adipose thermogenesis and metabolic homeostasis, as demonstrated in this study. Results: IEC-Exo can accumulate in adipose tissue. Compared with IEC-Exo derived from room temperature mice (RT IEC-Exo), Cold IEC-Exo significantly enhanced the thermogenesis of adipose. In vitro, Cold IEC-Exo directly stimulated thermogenesis in primary adipocytes by elevating oxygen consumption rate, proton leak, and fatty acid uptake, with no effect on glucose uptake. Small RNA sequencing identified miR-674-3p as a key mediator enriched in Cold IEC-Exo. miR-674-3p mimicry replicated Cold IEC-Exo effects, augmenting Ucp1 expression, mitochondrial uncoupling, and fatty acid utilization in adipocytes. Local overexpression of miR-674-3p in BAT and sWAT via AAV in vivo enhanced thermogenesis and attenuated diet-induced glucose intolerance. Conclusions: These findings establish that Cold IEC-Exo, via miR-674-3p transfer, drive adipose thermogenic activation and mitigate metabolic dysfunction, highlighting their therapeutic potential in obesity-related disorders.
Collapse
Affiliation(s)
- Xue Han
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Tiange Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yaxu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ziming Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Fangyu Shao
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yue Yin
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| |
Collapse
|
5
|
Emont MP, Essene AL, Gulko A, Bozadjieva-Kramer N, Jacobs C, Nagesh S, Seeley RJ, Tsai LT, Rosen ED. Semaglutide and bariatric surgery induce distinct changes in the composition of mouse white adipose tissue. Mol Metab 2025; 95:102126. [PMID: 40139440 PMCID: PMC11999362 DOI: 10.1016/j.molmet.2025.102126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Adipose tissue is a central player in energy balance and glucose homeostasis, expanding in the face of caloric overload in order to store energy safely. If caloric overload continues unabated, however, adipose tissue becomes dysfunctional, leading to systemic metabolic compromise in the form of insulin resistance and type 2 diabetes. Changes in adipose tissue during the development of metabolic disease are varied and complex, made all the more so by the heterogeneity of cell types within the tissue. Here we present detailed comparisons of atlases of murine WAT in the setting of diet-induced obesity, as well as after weight loss induced by either vertical sleeve gastrectomy (VSG) or treatment with the GLP-1 receptor agonist semaglutide. We focus on identifying populations of cells that return to a lean-like phenotype versus those that persist from the obese state, and examine pathways regulated in these cell types across conditions. These data provide a resource for the study of the cell type changes in WAT during weight loss, and paint a clearer picture of the differences between adipose tissue from lean animals that have never been obese, versus those that have.
Collapse
Affiliation(s)
- Margo P Emont
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Adam L Essene
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Anton Gulko
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Veterans Affairs Ann Arbor Healthcare System, Research Service, Ann Arbor, MI, USA
| | - Christopher Jacobs
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soumya Nagesh
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Linus T Tsai
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Park JS, Kim KS, Choi HJ. Glucagon-Like Peptide-1 and Hypothalamic Regulation of Satiation: Cognitive and Neural Insights from Human and Animal Studies. Diabetes Metab J 2025; 49:333-347. [PMID: 40367985 PMCID: PMC12086555 DOI: 10.4093/dmj.2025.0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have emerged as blockbuster drugs for treating metabolic diseases. Glucagon-like peptide-1 (GLP-1) plays a pivotal role in glucose homeostasis by enhancing insulin secretion, suppressing glucagon release, delaying gastric emptying, and acting on the central nervous system to regulate satiation and satiety. This review summarizes the discovery of GLP-1 and the development of GLP-1RAs, with a particular focus on their central mechanisms of action. Human neuroimaging studies demonstrate that GLP-1RAs influence brain activity during food cognition, supporting a role in pre-ingestive satiation. Animal studies on hypothalamic feed-forward regulation of hunger suggest that cognitive hypothalamic mechanisms may also contribute to satiation control. We highlight the brain mechanisms of GLP-1RA-induced satiation and satiety, including cognitive impacts, with an emphasis on animal studies of hypothalamic glucagon-like peptide-1 receptor (GLP-1R) and GLP-1R-expressing neurons. Actions in non-hypothalamic regions are also discussed. Additionally, we review emerging combination drugs and oral GLP-1RA formulations aimed at improving efficacy and patient adherence. In conclusion, the dorsomedial hypothalamus (DMH)-a key GLP-1RA target-mediates pre-ingestive cognitive satiation, while other hypothalamic GLP-1R neurons regulate diverse aspects of feeding behavior, offering potential therapeutic targets for obesity treatment.
Collapse
Affiliation(s)
- Joon Seok Park
- Department of Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyu Sik Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Hyung Jin Choi
- Department of Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Korea
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
7
|
Zhao X, Liu Y, Wang D, Li T, Xu Z, Li Z, Bai X, Wang Y. Role of GLP‑1 receptor agonists in sepsis and their therapeutic potential in sepsis‑induced muscle atrophy (Review). Int J Mol Med 2025; 55:74. [PMID: 40052580 PMCID: PMC11936484 DOI: 10.3892/ijmm.2025.5515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/17/2025] [Indexed: 03/27/2025] Open
Abstract
Sepsis‑induced myopathy (SIM) is a common complication in intensive care units, which is often associated with adverse outcomes, primarily manifested as skeletal muscle weakness and atrophy. Currently, the management of SIM focuses on prevention strategies, as effective therapeutic options remain elusive. Glucagon‑like peptide‑1 (GLP‑1) receptor agonists (GLP‑1RAs) have garnered attention as hypoglycemic and weight‑loss agents, with an increasing body of research focusing on the extrapancreatic effects of GLP‑1. In preclinical settings, GLP‑1RAs exert protective effects against sepsis‑related multiple organ dysfunction through anti‑inflammatory and antioxidant mechanisms. Based on the existing research, we hypothesized that GLP‑1RAs may serve potential protective roles in the repair and regeneration of skeletal muscle affected by sepsis. The present review aimed to explore the relationship between GLP‑1RAs and sepsis, as well as their impact on muscle atrophy‑related myopathy. Furthermore, the potential mechanisms and therapeutic benefits of GLP‑1RAs are discussed in the context of muscle atrophy induced by sepsis.
Collapse
Affiliation(s)
- Xuan Zhao
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dongfang Wang
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Tonghan Li
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhikai Xu
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhanfei Li
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiangjun Bai
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuchang Wang
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
8
|
Herz CT, Kulterer OC, Prager M, Marculescu R, Prager G, Kautzky-Willer A, Hacker M, Trajanoski S, Köfeler HC, Gallé B, Haug AR, Berry D, Kiefer FW. Bariatric surgery promotes recruitment of brown fat linked to alterations in the gut microbiota. Eur J Endocrinol 2025; 192:603-611. [PMID: 40366070 DOI: 10.1093/ejendo/lvaf081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/10/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVE The mechanisms of bariatric surgery-induced weight loss and metabolic improvements are still incompletely understood and reach beyond malabsorption or calorie restriction. We sought to investigate the effect of bariatric surgery on brown adipose tissue (BAT) activity and a potential connection with changes in energy metabolism, the gut microbiota, and short-chain fatty acid (SCFA) composition. METHODS We included 32 subjects (25 females) with morbid obesity and analyzed their metabolic profile, gut microbiota composition, circulating SCFAs, energy expenditure, and cold-induced BAT activity using [18F]Fluorodeoxyglucose-positron emission tomography-computed tomography before and up to 1 year after bariatric surgery. RESULTS Twelve months after surgery, the percentage of individuals with active BAT had increased from 28% to 53%. The BAT-negative (BATneg) individuals who had an adverse metabolic profile at baseline compared with subjects with active BAT (BATpos) showed a greater metabolic benefit after surgery. While no changes in overall gut bacterial diversity were observed between BATpos and BATneg, the abundance of 3 specific bacterial families, including Akkermansiaceae, Pasteurellaceae, and Carnobacteriaceae, was distinctly regulated between BAT groups. The bacterial genera most strongly increased in BATpos vs BATneg subjects were all positively correlated with BAT volume and BAT activity. Finally, circulating concentrations of the SCFAs acetate, butyrate, and propionate rose after bariatric surgery and were related to bacterial genera such as Akkermansia, Dialister, and Lachnospiraceae FCS020 group, all known SCFA producers. CONCLUSIONS Bariatric surgery helps recruit active BAT in individuals with obesity and is linked to distinct alterations in the gut microbiome and SCFA composition. TRIAL REGISTRATION NUMBER ClinicalTrials.gov (NCT03168009).
Collapse
Affiliation(s)
- Carsten T Herz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Oana C Kulterer
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Marlene Prager
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Gerhard Prager
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Alexandra Kautzky-Willer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Harald C Köfeler
- Core Facility Mass Spectrometry, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Birgit Gallé
- Core Facility Molecular Biology, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Alexander R Haug
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - David Berry
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Florian W Kiefer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Li B, Gao Y, Wang W, Zhu R, Yang X, Chen H, Wang X, Gu H. Epicardial Adipose Tissue and Extracellular Volume Fraction in Patients with Hypertrophic Cardiomyopathy: A Multi‑center Prognosis Study. Acad Radiol 2025:S1076-6332(25)00314-9. [PMID: 40268603 DOI: 10.1016/j.acra.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025]
Abstract
RATIONALE AND OBJECTIVES Epicardial adipose tissue (EAT) is thought to have a deleterious effect on the progression of myocardial disease; the extracellular volume (ECV) fraction has been validated by histology to correlate with adverse myocardial remodeling. The aim of this study was to investigate the prognostic value of EAT volume index (EATVI) and ECV in patients with hypertrophic cardiomyopathy (HCM). MATERIALS AND METHODS ECV and EAT were measured using cardiac magnetic resonance (CMR) imaging in 201 subjects with HCM. All patients were followed up prospectively. Major adverse cardiovascular events (MACEs) were categorized into primary and secondary endpoint events. The primary endpoint was a composite of cardiac death, heart transplant, and cardiopulmonary resuscitation following syncope. The secondary endpoint was defined as rehospitalization for heart failure (HF). RESULTS After 26±16 months of follow-up, 43 patients experienced MACEs (14 patients experienced a primary endpoint, and 29 patients experienced a secondary endpoint). Patients suffering from MACEs had significantly higher ECV and EATVI (p<0.001). After adjustment for body mass index (BMI), EATVI showed a significant correlation with ECV (r=0.424, p<0.001) among HCM patients. In the Kaplan-Meier analysis, the incidence of MACE was significantly higher in patients with increased ECV (p<0.001) and higher EATVI (p<0.001). In multivariate Cox regression analysis, ECV (HR=1.12, p<0.001) and EATVI (HR=1.31, p<0.001) were significantly associated with MACEs. CONCLUSION In patients with HCM, ECV and EATVI measured by CMR are strong predictors of MACEs and improve risk stratification. CLINICAL IMPLICATION Extracellular volume fraction and epicardial adipose tissue are associated with major adverse cardiovascular events in patients with hypertrophic cardiomyopathy and can improve their risk stratification.
Collapse
Affiliation(s)
- Bowen Li
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250117, China (B.L., R.Z., X.Y., H.C.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.)
| | - Yan Gao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.Y., X.W.)
| | - Wenxian Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.); School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Yantai, Shandong 264003, PR China (W.W., X.Y.)
| | - Runze Zhu
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250117, China (B.L., R.Z., X.Y., H.C.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.)
| | - Xueqiao Yang
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250117, China (B.L., R.Z., X.Y., H.C.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.Y., X.W.); School of Medical Imaging, Binzhou Medical University, No. 346 Guanhai Road, Yantai, Shandong 264003, PR China (W.W., X.Y.)
| | - Huiyu Chen
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250117, China (B.L., R.Z., X.Y., H.C.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.)
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.Y., X.W.)
| | - Hui Gu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China (B.L., Y.G., W.W., R.Z., X.Y., H.C., X.W., H.G.).
| |
Collapse
|
10
|
Janssen-Telders C, Eringa EC, de Groot JR, de Man FS, Handoko ML. The role of epicardial adipose tissue remodelling in heart failure with preserved ejection fraction. Cardiovasc Res 2025:cvaf056. [PMID: 40238568 DOI: 10.1093/cvr/cvaf056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/12/2024] [Accepted: 01/22/2025] [Indexed: 04/18/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a growing global health problem characterized by high morbidity and mortality, with limited effective therapies available. Obesity significantly influences haemodynamic and structural changes in the myocardium and vasculature, primarily through the accumulation and action of visceral adipose tissue. Particularly, epicardial adipose tissue (EAT) contributes to HFpEF through inflammation and lipotoxic infiltration of the myocardium. However, the precise signalling pathways leading to diastolic stiffness in HFpEF require further elucidation. This review explores the dynamic role of EAT in health and disease. Drawing upon insights from studies in other conditions, we discuss potential EAT-mediated inflammatory pathways in HFpEF and how they may contribute to functional and structural myocardial and endothelial derangements, including intramyocardial lipid infiltration, fibrosis, endothelial dysfunction, cardiomyocyte stiffening, and left ventricular hypertrophy. Lastly, we propose potential targets for novel therapeutic avenues.
Collapse
Affiliation(s)
- Carolina Janssen-Telders
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Etto C Eringa
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Department of Physiology, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht UMC, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Joris R de Groot
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Frances S de Man
- Amsterdam Cardiovascular Sciences, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht UMC, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - M Louis Handoko
- Department of Cardiology Amsterdam UMC, Heart Centre, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Pulmonology, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
11
|
Xu Z, Wen S, Dong M, Zhou L. Targeting central pathway of Glucose-Dependent Insulinotropic Polypeptide, Glucagon and Glucagon-like Peptide-1 for metabolic regulation in obesity and type 2 diabetes. Diabetes Obes Metab 2025; 27:1660-1675. [PMID: 39723473 DOI: 10.1111/dom.16146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Obesity and type 2 diabetes are significant public health challenges that greatly impact global well-being. The development of effective therapeutic strategies has become more and more concentrated on the central nervous system and metabolic regulation. The primary pharmaceutical interventions for the treatment of obesity and uncontrolled hyperglycemia are now generally considered to be incretin-based anti-diabetic treatments, particularly glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide receptor agonists. This is a result of their substantial influence on the central nervous system and the consequent effects on energy balance and glucose regulation. It is increasingly crucial to understand the neural pathways of these pharmaceuticals. The purpose of this review is to compile and present the most recent central pathways regarding glucagon-like peptide-1, glucose-dependent insulinotropic polypeptide and glucagon receptors, with a particular emphasis on central metabolic regulation.
Collapse
Affiliation(s)
- Zhimin Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- Fudan Zhangjiang Institute, Fudan University, Shanghai, China
| | - Meiyuan Dong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
12
|
Reiss AB, Gulkarov S, Lau R, Klek SP, Srivastava A, Renna HA, De Leon J. Weight Reduction with GLP-1 Agonists and Paths for Discontinuation While Maintaining Weight Loss. Biomolecules 2025; 15:408. [PMID: 40149944 PMCID: PMC11940170 DOI: 10.3390/biom15030408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Worldwide, nearly 40% of adults are overweight and 13% are obese. Health consequences of excess weight include cardiovascular diseases, type 2 diabetes, dyslipidemia, and increased mortality. Treating obesity is challenging and calorie restriction often leads to rebound weight gain. Treatments such as bariatric surgery create hesitancy among patients due to their invasiveness. GLP-1 medications have revolutionized weight loss and can reduce body weight in obese patients by between 15% and 25% on average after about 1 year. Their mode of action is to mimic the endogenous GLP-1, an intestinal hormone that regulates glucose metabolism and satiety. However, GLP-1 drugs carry known risks and, since their use for weight loss is recent, may carry unforeseen risks as well. They carry a boxed warning for people with a personal or family history of medullary thyroid carcinoma or multiple endocrine neoplasia syndrome type 2. Gastrointestinal adverse events (nausea, vomiting, diarrhea) are fairly common while pancreatitis and intestinal obstruction are rarer. There may be a loss of lean body mass as well as premature facial aging. A significant disadvantage of using these medications is the high rate of weight regain when they are discontinued. Achieving success with pharmacologic treatment and then weaning to avoid future negative effects would be ideal.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (R.L.); (S.P.K.); (J.D.L.)
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (A.S.); (H.A.R.)
| | - Shelly Gulkarov
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (A.S.); (H.A.R.)
| | - Raymond Lau
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (R.L.); (S.P.K.); (J.D.L.)
| | - Stanislaw P. Klek
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (R.L.); (S.P.K.); (J.D.L.)
| | - Ankita Srivastava
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (A.S.); (H.A.R.)
| | - Heather A. Renna
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (A.S.); (H.A.R.)
| | - Joshua De Leon
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (R.L.); (S.P.K.); (J.D.L.)
| |
Collapse
|
13
|
Rodríguez-Díaz A, Diéguez C, López M, Freire-Agulleiro Ó. FAcTs on fire: Exploring thermogenesis. ADVANCES IN GENETICS 2025; 113:172-198. [PMID: 40409797 DOI: 10.1016/bs.adgen.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Thermoregulation is a fundamental biological process that allows birds and mammals to maintain a stable internal temperature despite environmental fluctuations, a mechanism shaped by millions of years of evolution. Non-shivering thermogenesis (NST), primarily driven by brown adipose tissue (BAT), plays a central role in thermoregulation by not only helping maintain energy homeostasis but also influencing broader metabolic and physiological processes. Recent research has revealed that BAT thermogenesis is regulated by peripheral hormones and at a central level, with key hypothalamic energy-sensing pathways-such as AMP-activated protein kinase (AMPK) and endoplasmic reticulum (ER) stress-playing critical roles. Beyond its metabolic functions, BAT and NST have emerged as important contributors to tumor biology, offering novel therapeutic strategies for metabolic and oncological diseases. This review explores the intricate mechanisms underpinning NST, including UCP1-dependent thermogenesis and alternative pathways such as creatine cycling, calcium-dependent thermogenesis, and lipid cycling. Emerging evidence further highlights BAT's potential in to modulate tumor metabolism, with pharmacological and genetic approaches showing promise in reshaping the tumor microenvironment. This growing body of knowledge offers exciting prospects for targeting BAT thermogenesis in treating obesity and other metabolic diseases.
Collapse
Affiliation(s)
- Amanda Rodríguez-Díaz
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | - Óscar Freire-Agulleiro
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.
| |
Collapse
|
14
|
Gaggini M, Sabatino L, Suman AF, Chatzianagnostou K, Vassalle C. Insights into the Roles of GLP-1, DPP-4, and SGLT2 at the Crossroads of Cardiovascular, Renal, and Metabolic Pathophysiology. Cells 2025; 14:387. [PMID: 40072115 PMCID: PMC11898734 DOI: 10.3390/cells14050387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025] Open
Abstract
In recent years, new drugs for the treatment of type 2 diabetes (T2D) have been proposed, including glucagon-like peptide 1 (GLP-1) agonists or sodium-glucose cotransporter 2 (SGLT2) inhibitors and dipeptidyl peptidase-4 (DPP-4) inhibitors. Over time, some of these agents (in particular, GLP-1 agonists and SGLT2 inhibitors), which were initially developed for their glucose-lowering actions, have demonstrated significant beneficial pleiotropic effects, thus expanding their potential therapeutic applications. This review aims to discuss the mechanisms, pleiotropic effects, and therapeutic potential of GLP-1, DPP-4, and SGLT2, with a particular focus on their cardiorenal benefits beyond glycemic control.
Collapse
Affiliation(s)
- Melania Gaggini
- Institute of Clinical Physiology, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy; (M.G.); (L.S.)
| | - Laura Sabatino
- Institute of Clinical Physiology, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy; (M.G.); (L.S.)
| | - Adrian Florentin Suman
- Institute of Clinical Physiology, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy; (M.G.); (L.S.)
| | | | - Cristina Vassalle
- Fondazione CNR-Regione Toscana G Monasterio, Via G. Moruzzi 1, 56124 Pisa, Italy;
| |
Collapse
|
15
|
Pahlavani M, Pham K, Kalupahana NS, Morovati A, Ramalingam L, Abidi H, Kiridana V, Moustaid-Moussa N. Thermogenic adipose tissues: Promising therapeutic targets for metabolic diseases. J Nutr Biochem 2025; 137:109832. [PMID: 39653156 DOI: 10.1016/j.jnutbio.2024.109832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
The ongoing increase in the prevalence of obesity and its comorbidities such as cardiovascular disease, type 2 diabetes (T2D) and dyslipidemia warrants discovery of novel therapeutic options for these metabolic diseases. Obesity is characterized by white adipose tissue expansion due to chronic positive energy balance as a result of excessive energy intake and/or reduced energy expenditure. Despite various efforts to prevent or reduce obesity including lifestyle and behavioral interventions, surgical weight reduction approaches and pharmacological methods, there has been limited success in significantly reducing obesity prevalence. Recent research has shown that thermogenic adipocyte (brown and beige) activation or formation, respectively, could potentially act as a therapeutic strategy to ameliorate obesity and its related disorders. This can be achieved through the ability of these thermogenic cells to enhance energy expenditure and regulate circulating levels of glucose and lipids. Thus, unraveling the molecular mechanisms behind the formation and activation of brown and beige adipocytes holds the potential for probable therapeutic paths to combat obesity. In this review, we provide a comprehensive update on the development and regulation of different adipose tissue types. We also emphasize recent interventions in harnessing therapeutic potential of thermogenic adipocytes by bioactive compounds and new pharmacological anti-obesity agents.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Sciences, Texas Woman's University, Dallas, Texas, USA
| | - Kenneth Pham
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Ashti Morovati
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Studies, Syracuse University, Syracuse, New York, USA
| | - Hussain Abidi
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Vasana Kiridana
- Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Institute for One Health Innovation, Texas Tech University and Texas Tech Health Sciences Center, Lubbock, Texas, USA.
| |
Collapse
|
16
|
Li W, Zhang X, Song J, Yang L, Wang D, Yuan G, Zhao L. Mechanistic insights into GLP-1 receptor agonist-induced weight loss through ceRNA network analysis. Genomics 2025; 117:110988. [PMID: 39761765 DOI: 10.1016/j.ygeno.2025.110988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND GLP-1 receptor agonists (GLP-1RA) have been extensively utilized in the management of body weight in individuals with obesity. Circular RNA (circRNA), a class of covalently closed RNA molecules, has garnered increasing attention for its potential role in the pathogenesis of obesity. However, the specific mechanisms through which circRNA contributes to GLP-1RA-induced weight loss remains elusive. METHODS High-throughput sequencing analyzed epididymal adipose tissue from obese mice under high-fat, and GLP-1RA intervention (600 μg/kg/d). The functions of differentially expressed (DE) genes were enriched and analyzed. The circRNA-miRNA-mRNA interaction network was constructed in Cytoscape, and KEGG pathway gene enrichment was validated via western blotting. RESULTS A total of 644 DEcircRNAs, 186 DEmiRNAs, and 3474 DEmRNAs were identified. Based on ceRNA score calculations, network diagrams were constructed. Gene Ontology (GO) analysis revealed that DERNAs were linked to lipid and fatty acid metabolism. DE genes within ceRNA pairs were enriched in lipid metabolism pathways, especially the PI3K-Akt and AMPK signaling pathways. GLP-1RA induced the phosphorylation of AKT and AMPK, which subsequently led to a reduction of SREBP-1, ACC, and FAS. CONCLUSION GLP-1RA might activate PI3K-Akt and AMPK signaling pathways to combat obesity through the ceRNA network of circRNAs.
Collapse
Affiliation(s)
- Wenxin Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Xinyu Zhang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang 212000, Jiangsu, China; Department of Endocrinology, the Eighth People's Hospital of Wuxi, Wuxi 214000, Jiangsu, China
| | - Jiamin Song
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Ling Yang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Dong Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang 212000, Jiangsu, China.
| | - Li Zhao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University, Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang 212000, Jiangsu, China.
| |
Collapse
|
17
|
Argyrakopoulou G, Gitsi E, Konstantinidou SK, Kokkinos A. The effect of obesity pharmacotherapy on body composition, including muscle mass. Int J Obes (Lond) 2025; 49:381-387. [PMID: 38745020 DOI: 10.1038/s41366-024-01533-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
Obesity pharmacotherapy represents a promising approach to treating obesity and may provide benefits beyond weight loss alone. Maintaining or even increasing muscle mass during weight loss is important to overall health, metabolic function and weight loss maintenance. Drugs such as liraglutide, semaglutide, tirzepatide, and naltrexone/bupropion have shown significant weight loss effects, and emerging evidence suggests they may also have effects on body composition, particularly a positive influence on muscle mass. However, further research is needed to fully understand the mechanism of action of these drugs and their effects on muscle mass. Clinicians should consider these factors when developing an obesity treatment plan for an individual patient.
Collapse
Affiliation(s)
| | - Evdoxia Gitsi
- Diabetes and Obesity Unit, Athens Medical Center, 15125, Athens, Greece
| | - Sofia K Konstantinidou
- Diabetes and Obesity Unit, Athens Medical Center, 15125, Athens, Greece
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
18
|
Hurtado-Carneiro V, Juan-Arevalo Y, Flores CN, Herrero-De-Dios C, Perez-Garcia A, Contreras C, Lopez M, Alvarez E, Sanz C. Enhanced thermogenesis in PAS Kinase-deficient male mice. Biochem Pharmacol 2025; 233:116757. [PMID: 39824466 DOI: 10.1016/j.bcp.2025.116757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/15/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
PAS domain-containing serine/threonine-protein kinase (PASK) is a nutrient and energy sensor regulated by fasting/refeeding conditions in hypothalamic areas involved in controlling energy balance. In this sense, PASK plays a role in coordinating the activation/inactivation of AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) in response to fasting. PASK deficiency protects against the development of diet-induced obesity. This has prompted an investigation into the potential role of PASK on energy expenditure through thermogenesis in adipose tissue. Our results indicate that PASK-deficient male mice exhibited higher brown adipose tissue (BAT) thermogenic activity and heat production. The inhibition of PASK function induces the expression of Uncoupling Protein 1 (UCP1) and the adipogenic marker peroxisome proliferator-activated receptor gamma (PPARγ) in BAT. In addition, PASK deficiency promotes the expression of UCP1 and other browning markers such as PR/SET Domain 16 (PRDM16) in inguinal white adipose tissue (WAT). PASK-deficient mice record an enhanced thermogenic response, even under stimuli such as β-3adrenergic receptor agonist or cold. This evidence reveals PASK as a new mechanism modulating BAT thermogenesis.
Collapse
Affiliation(s)
- Veronica Hurtado-Carneiro
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Spain; Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Madrid, Spain.
| | - Yolanda Juan-Arevalo
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Spain; Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| | - Cinthya N Flores
- Department of Physiology, Faculty of Medicine, Complutense University of Madrid, Spain; Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| | - Carmen Herrero-De-Dios
- Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| | - Ana Perez-Garcia
- Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| | - Cristina Contreras
- Department of Physiology, Faculty of Pharmacy, Complutense University of Madrid, Spain; NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Lopez
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Elvira Alvarez
- Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Madrid, Spain
| | - Carmen Sanz
- Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Madrid, Spain; Department of Cell Biology, Faculty of Medicine, Complutense University of Madrid, Spain
| |
Collapse
|
19
|
Zhang Z, Liu S, Xian J, Zhang Y, Zhang C, Wang Z, Deng H, Feng J, Yao L. Effect of Hypoglycemic Drugs on Patients with Heart Failure with or without T2DM: A Bayesian Network Meta-analysis. Rev Cardiovasc Med 2025; 26:26154. [PMID: 40160590 PMCID: PMC11951290 DOI: 10.31083/rcm26154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 04/02/2025] Open
Abstract
Background Anti-diabetic drugs have been noted to have a cardioprotective effect in patients with diabetes and heart failure (HF). The purpose of this study was to perform a Bayesian network meta-analysis to evaluate the impact of various anti-diabetic drugs on the prognosis of HF patients with and without diabetes. Methods We searched PubMed, Embase, Cochrane, and Web of Science for randomized controlled trials (RCTs) published before November 2024 that investigated the use of anti-diabetic medications in patients with HF. Primary outcomes included re-admission due to HF, all-cause death, cardiovascular death, serum N-terminal pro-brain natriuretic peptide (NTpro-BNP) levels, and left ventricular ejection fraction (LVEF). A Bayesian network meta-analysis was used to compare the effectiveness of different anti-diabetic drugs. Results A total of 33 RCTs involving 29,888 patients were included. Sotagliflozin was the most effective in reducing the risk of re-admission due to HF and all-cause death, with a cumulative probability of 0.84 and 0.83, respectively. Liraglutide reduced the risk of cardiovascular death in HF patients with a cumulative probability of 0.97 and had the best efficacy in reducing NTpro-BNP levels with a cumulative probability of 0.69. Empagliflozin was best in improving LVEF in HF patients, with a cumulative probability of 0.69. Conclusions This Bayesian network meta-analysis demonstrates that sotagliflozin may be the best option for HF patients with and without diabetes. However, due to the small number of articles in this study, our results must be treated cautiously. Subsequently, there is an urgent need for more high-quality studies to validate our findings.
Collapse
Affiliation(s)
- Zhaolun Zhang
- Department of Cardiovascular Medicine, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Siqi Liu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Jiawen Xian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yali Zhang
- Department of Cardiovascular Medicine, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Chunyu Zhang
- Department of Cardiovascular Medicine, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Zhiyuan Wang
- Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Hongmei Deng
- Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Jian Feng
- Department of Cardiovascular Medicine, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Lei Yao
- Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072 Chengdu, Sichuan, China
- The Center for Medical Genetics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072 Chengdu, Sichuan, China
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072 Chengdu, Sichuan, China
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, 610072 Chengdu, Sichuan, China
- Medical Experiment Center, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
20
|
Yu H, Feng N, Zhong W, Han Y, Cheng Y, Zhang Z, Wang Y, Gao P, Huang R, Zhang C, Liu Z, Dong J, He Z, Lai H, Shen Z, Zhai Q. Nmnat2 deficiency in the arcuate nucleus or paraventricular nucleus induces Sarm1-independent neuron loss and liraglutide-reversible obesity. FASEB J 2025; 39:e70400. [PMID: 39964232 DOI: 10.1096/fj.202402546r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 05/10/2025]
Abstract
Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) plays an important role in maintaining axon integrity, and the arcuate nucleus (ARC), and paraventricular nucleus (PVN) are crucial nuclei in the control of energy balance. However, the effect of Nmnat2 deficiency in ARC and PVN is still unclear. Nmnat2 loxP/loxP or Nmnat2 loxP/loxP , Sarm1 -/- mice were bilaterally injected with AAV-CMV-GFP-Cre once into the ARC, PVN, or lateral parabrachial nucleus (LPBN) to obtain Nmnat2 ARC-/- , Nmnat2 PVN-/- , Nmnat2 LPBN-/- , Nmnat2 ARC-/- , SKO, Nmnat2 PVN-/- , SKO, or Nmnat2 LPBN-/- , SKO mice. Syn1-Cre mice were bilaterally injected with AAV-EF1a-flex-taCasp3-TEVp once into the ARC or PVN to specifically induce neuron loss. Metabolic changes were measured in the mice intraperitoneally injected with or without liraglutide, a glucagon-like peptide-1 (GLP-1) analog. Neuron loss and neuron activation were monitored by immunofluorescence. Deletion of Nmnat2 in ARC or PVN of mice leads to neuron loss, increased food intake, and obesity in a Sarm1-independent manner. Intraperitoneal injection of liraglutide activates neurons in PVN and LPBN, and attenuates hyperphagia and obesity induced by Nmnat2 deletion or apoptosis of Syn1-positive neurons in ARC or PVN, but has no significant effect on neuron loss. Nmnat2 deficiency in LPBN leads to death within 2 weeks, which can be markedly rescued by Sarm1 deficiency. These data show that deletion of Nmnat2 in ARC or PVN in adult mice leads to Sarm1-independent neuron loss, and liraglutide-reversible hyperphagia and obesity. These findings also elucidate the integrated role of ARC or PVN for downregulating food intake, the requirement of LPBN for survival, and the ARC- or PVN-independent effect of GLP-1 on food intake.
Collapse
Affiliation(s)
- Huimin Yu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ning Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wuling Zhong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yumo Han
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yalan Cheng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhentong Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingqi Wang
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peidong Gao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Huang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Cong Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zongyang Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jieya Dong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhishui He
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hejin Lai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ziru Shen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
21
|
Subedi L, Bamjan AD, Phuyal S, Shim JH, Cho SS, Seo JB, Chang KY, Byun Y, Kweon S, Park JW. An oral liraglutide nanomicelle formulation conferring reduced insulin-resistance and long-term hypoglycemic and lipid metabolic benefits. J Control Release 2025; 378:637-655. [PMID: 39709071 DOI: 10.1016/j.jconrel.2024.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/20/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Type 2 diabetes is a chronic disease characterized by insulin resistance and often worsened by obesity. Effective management involves the use of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) to assist with glycemic control and weight management. However, these drugs must be administered subcutaneously due to their low oral bioavailability. We developed an oral liraglutide (LRG) formulation by electrostatic complexation of GLP-1 RA with bile acid derivatives and nanomicelle (NM) formation, with non-ionic surfactant n-dodecyl-β-d-maltoside (DDM). The optimized formulation, LDD[1:2:4]-NM, had a mean particle size of 75.9 ± 5.60 nm and a permeability 1347 % higher than that of unformulated LRG when tested in Caco-2/HT29-MTX-E12 cell monolayers. In rats, oral bioavailability was 4.63-fold higher than that of unformulated LRG (1.11 ± 0.20 % vs. 5.14 ± 0.63 %). The absorption mechanism included clathrin-mediated endocytosis, macropinocytosis, and an ASBT-mediated pathway. A 12-week oral treatment consisting of a daily dose of 20 mg LDD[1:2:4]-NM/kg significantly reduced glycohemoglobin levels, a marker of diabetic control, and the HOMA-IR index, a marker of insulin resistance. The weight of epididymal and inguinal white adipose tissue and brown adipose tissue (BAT) was also reduced. Moreover, LDD[1:2:4]-NM had a greater impact on BAT activation, pro-inflammatory gene expression, and lipid metabolism than subcutaneous LRG. This study showed that an oral NM formulation can efficiently deliver LRG. Long-term treatment led to improved hyperglycemic effects, insulin resistance, and modulated lipid metabolism. LDD[1:2:4]-NM is thus a promising oral therapeutic option for the management of type 2 diabetes, potentially transforming treatment paradigms based on the availability of a more convenient administration route.
Collapse
Affiliation(s)
- Laxman Subedi
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Arjun Dhwoj Bamjan
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Susmita Phuyal
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Seung-Sik Cho
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jong Bae Seo
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | | | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea.
| |
Collapse
|
22
|
Schooling CM, Yang G, Soliman GA, Leung GM. A Hypothesis That Glucagon-like Peptide-1 Receptor Agonists Exert Immediate and Multifaceted Effects by Activating Adenosine Monophosphate-Activate Protein Kinase (AMPK). Life (Basel) 2025; 15:253. [PMID: 40003662 PMCID: PMC11857512 DOI: 10.3390/life15020253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) reduce bodyweight and blood glucose. Extensive evidence from randomized controlled trials has indicated that GLP-1RAs have benefits well beyond weight loss and glucose control, extending from reductions in cardiovascular mortality to reductions in prostate cancer risk. Notably, some benefits of GLP-1RAs for the cardiovascular-kidney-metabolic (CKM) system arise before weight loss occurs for reasons that are not entirely clear but are key to patient care and drug development. Here, we hypothesize that GLP-1RAs act by inducing calorie restriction and by activating adenosine monophosphate-activated protein kinase (AMPK), which not only provides an explanation for the unique effectiveness of GLP-1RAs but also indicates a common mechanism shared by effective CKM therapies, including salicylates, metformin, statins, healthy diet, and physical activity. Whether AMPK activation is obligatory for effective CKM therapies should be considered. As such, we propose a mechanism of action for GLP-1RAs and explain how it provides an overarching framework for identifying means of preventing and treating cardiovascular, kidney, metabolic and related diseases, as well as informing the complementary question as to the components of a healthy lifestyle.
Collapse
Affiliation(s)
- C. Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (G.Y.)
- School of Public Health and Health Policy, City University of New York, Graduate School of Public Health and Health Policy, 55 W 125th St, New York, NY 10027, USA;
| | - Guoyi Yang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (G.Y.)
| | - Ghada A. Soliman
- School of Public Health and Health Policy, City University of New York, Graduate School of Public Health and Health Policy, 55 W 125th St, New York, NY 10027, USA;
| | - Gabriel M. Leung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (G.Y.)
| |
Collapse
|
23
|
Tiezzi M, Vieceli Dalla Sega F, Gentileschi P, Campanelli M, Benavoli D, Tremoli E. Effects of Weight Loss on Endothelium and Vascular Homeostasis: Impact on Cardiovascular Risk. Biomedicines 2025; 13:381. [PMID: 40002792 PMCID: PMC11853214 DOI: 10.3390/biomedicines13020381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Available knowledge shows that obesity is associated with an impaired endothelial function and an increase in cardiovascular risk, but the mechanisms of this association are not yet fully understood. Adipose tissue dysfunction, adipocytokines production, along with systemic inflammation and associated comorbidities (e.g., diabetes and hypertension), are regarded as the primary physiological and pathological factors. Various strategies are now available for the control of excess body weight. Dietary regimens alone, or in association with bariatric surgery when indicated, are now widely used. Of particular interest is the understanding of the effect of these interventions on endothelial homeostasis in relation to cardiovascular health. Substantial weight loss resulting from both diet and bariatric surgery decreases circulating biomarkers and improves endothelial function. Extensive clinical trials and meta-analyses show that bariatric surgery (particularly gastric bypass) has more substantial and long-lasting effect on weight loss and glucose regulation, as well as on distinct circulating biomarkers of cardiovascular risk. This review summarizes the current understanding of the distinct effects of diet-induced and surgery-induced weight loss on endothelial function, focusing on the key mechanisms involved in these effects.
Collapse
Affiliation(s)
- Margherita Tiezzi
- Dipartimento Cardiovascolare, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy;
| | | | - Paolo Gentileschi
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
- Dipartimento di Scienze Chirurgiche, Università di Roma Tor Vergata, 00133 Roma, Italy
| | - Michela Campanelli
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
| | - Domenico Benavoli
- Dipartimento di Chirurgia Bariatrica e Metabolica, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy; (P.G.); (M.C.); (D.B.)
| | - Elena Tremoli
- Dipartimento Cardiovascolare, Maria Cecilia Hospital GVM Care and Research, 48033 Cotignola, Italy;
| |
Collapse
|
24
|
Duran M, Willis JR, Dalvi N, Fokakis Z, Virkus SA, Hardaway JA. Integration of Glucagon-Like Peptide 1 Receptor Actions Through the Central Amygdala. Endocrinology 2025; 166:bqaf019. [PMID: 39888375 PMCID: PMC11850305 DOI: 10.1210/endocr/bqaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/31/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
Understanding the detailed mechanism of action of glucagon-like peptide 1 receptor (GLP-1R) agonists on distinct topographic and genetically defined brain circuits is critical for improving the efficacy and mitigating adverse side effects of these compounds. In this mini-review, we propose that the central nucleus of the amygdala (CeA) is a critical mediator of GLP-1R agonist-driven hypophagia. Here, we review the extant literature demonstrating CeA activation via GLP-1R agonists across multiple species and through multiple routes of administration. The precise role of GLP-1Rs within the CeA is unclear but the site-specific GLP-1Rs may mediate distinct behavioral and physiological hallmarks of GLP-1R agonists on food intake. Thus, we propose important novel directions and methods to test the role of the CeA in mediating GLP-1R actions.
Collapse
Affiliation(s)
- Miguel Duran
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer R Willis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nilay Dalvi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zoe Fokakis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sonja A Virkus
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - J Andrew Hardaway
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
25
|
Hankir MK, Lutz TA. Novel neural pathways targeted by GLP-1R agonists and bariatric surgery. Pflugers Arch 2025; 477:171-185. [PMID: 39644359 PMCID: PMC11761532 DOI: 10.1007/s00424-024-03047-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
The glucagon-like peptide 1 receptor (GLP-1R) agonist semaglutide has revolutionized the treatment of obesity, with other gut hormone-based drugs lined up that show even greater weight-lowering ability in obese patients. Nevertheless, bariatric surgery remains the mainstay treatment for severe obesity and achieves unparalleled weight loss that generally stands the test of time. While their underlying mechanisms of action remain incompletely understood, it is clear that the common denominator between GLP-1R agonists and bariatric surgery is that they suppress food intake by targeting the brain. In this Review, we highlight recent preclinical studies using contemporary neuroscientific techniques that provide novel concepts in the neural control of food intake and body weight with reference to endogenous GLP-1, GLP-1R agonists, and bariatric surgery. We start in the periphery with vagal, intestinofugal, and spinal sensory nerves and then progress through the brainstem up to the hypothalamus and finish at non-canonical brain feeding centers such as the zona incerta and lateral septum. Further defining the commonalities and differences between GLP-1R agonists and bariatric surgery in terms of how they target the brain may not only help bridge the gap between pharmacological and surgical interventions for weight loss but also provide a neural basis for their combined use when each individually fails.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
26
|
Krieger JP, Daniels D, Lee S, Mastitskaya S, Langhans W. Glucagon-Like Peptide-1 Links Ingestion, Homeostasis, and the Heart. Compr Physiol 2025; 15:e7. [PMID: 39887844 PMCID: PMC11790259 DOI: 10.1002/cph4.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 02/01/2025]
Abstract
Glucagon-like peptide-1 (GLP-1), a hormone released from enteroendocrine cells in the distal small and large intestines in response to nutrients and other stimuli, not only controls eating and insulin release, but is also involved in drinking control as well as renal and cardiovascular functions. Moreover, GLP-1 functions as a central nervous system peptide transmitter, produced by preproglucagon (PPG) neurons in the hindbrain. Intestinal GLP-1 inhibits eating by activating vagal sensory neurons directly, via GLP-1 receptors (GLP-1Rs), but presumably also indirectly, by triggering the release of serotonin from enterochromaffin cells. GLP-1 enhances glucose-dependent insulin release via a vago-vagal reflex and by direct action on beta cells. Finally, intestinal GLP-1 acts on the kidneys to modulate electrolyte and water movements, and on the heart, where it provides numerous benefits, including anti-inflammatory, antiatherogenic, and vasodilatory effects, as well as protection against ischemia/reperfusion injury and arrhythmias. Hindbrain PPG neurons receive multiple inputs and project to many GLP-1R-expressing brain areas involved in reward, autonomic functions, and stress. PPG neuron-derived GLP-1 is involved in the termination of large meals and is implicated in the inhibition of water intake. This review details GLP-1's roles in these interconnected systems, highlighting recent findings and unresolved issues, and integrating them to discuss the physiological and pathological relevance of endogenous GLP-1 in coordinating these functions. As eating poses significant threats to metabolic, fluid, and immune homeostasis, the body needs mechanisms to mitigate these challenges while sustaining essential nutrient intake. Endogenous GLP-1 plays a crucial role in this "ingestive homeostasis."
Collapse
Affiliation(s)
- Jean-Philippe Krieger
- Jean-Philippe Krieger, Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Winterthurerstr. 260, 8057 Zurich
| | - Derek Daniels
- Department of Biological Sciences and the Center for Ingestive Behavior Research, University at Buffalo, the State University of New York, Buffalo NY 14260 USA
| | - Shin Lee
- Shin J. Lee, Neurimmune AG, Wagistrasse 18, 8952 Schlieren, Switzerland
| | - Svetlana Mastitskaya
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Dept. of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
27
|
Shimizu H, Miyamoto J, Hisa K, Ohue-Kitano R, Takada H, Yamano M, Nishida A, Sasahara D, Masujima Y, Watanabe K, Nishikawa S, Takahashi S, Ikeda T, Nakajima Y, Yoshida N, Matsuzaki C, Kageyama T, Hayashi I, Matsuki A, Akashi R, Kitahama S, Ueyama M, Murakami T, Inuki S, Irie J, Satoh-Asahara N, Toju H, Mori H, Nakaoka S, Yamashita T, Toyoda A, Yamamoto K, Ohno H, Katayama T, Itoh H, Kimura I. Sucrose-preferring gut microbes prevent host obesity by producing exopolysaccharides. Nat Commun 2025; 16:1145. [PMID: 39880823 PMCID: PMC11779931 DOI: 10.1038/s41467-025-56470-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025] Open
Abstract
Commensal bacteria affect host health by producing various metabolites from dietary carbohydrates via bacterial glycometabolism; however, the underlying mechanism of action remains unclear. Here, we identified Streptococcus salivarius as a unique anti-obesity commensal bacterium. We found that S. salivarius may prevent host obesity caused by excess sucrose intake via the exopolysaccharide (EPS) -short-chain fatty acid (SCFA) -carbohydrate metabolic axis in male mice. Healthy human donor-derived S. salivarius produced high EPS levels from sucrose but not from other sugars. S. salivarius abundance was significantly decreased in human donors with obesity compared with that in healthy donors, and the EPS-SCFA bacterial carbohydrate metabolic process was attenuated. Our findings reveal an important mechanism by which host-commensal interactions in glycometabolism affect energy regulation, suggesting an approach for preventing lifestyle-related diseases via prebiotics and probiotics by targeting bacteria and EPS metabolites.
Collapse
Affiliation(s)
- Hidenori Shimizu
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
- Noster Inc., Kamiueno, Muko-shi, Kyoto, Japan
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan
| | - Keiko Hisa
- Noster Inc., Kamiueno, Muko-shi, Kyoto, Japan
| | - Ryuji Ohue-Kitano
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Hiromi Takada
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Mayu Yamano
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Akari Nishida
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Daiki Sasahara
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
- Noster Inc., Kamiueno, Muko-shi, Kyoto, Japan
| | - Yuki Masujima
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Keita Watanabe
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Shota Nishikawa
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Sakura Takahashi
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Takako Ikeda
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yuya Nakajima
- Department of Endocrinology, Metabolism and Nephrology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Naofumi Yoshida
- Department of Advanced Medical Technologies, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Chiaki Matsuzaki
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi-shi, Ishikawa, Japan
| | - Takuya Kageyama
- Center for Ecological Research, Kyoto University, Otsu-shi, Shiga, Japan
| | - Ibuki Hayashi
- Laboratory of Ecosystems and Coevolution, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Akari Matsuki
- Laboratory of Mathematical Biology, Faculty of Advanced Life Science, Hokkaido University, Sapporo-shi, Hokkaido, Japan
- The Abdus Salam International Centre for Theoretical Physics (ICTP), Trieste, Italy
| | - Ryo Akashi
- Laboratory of Mathematical Biology, Faculty of Advanced Life Science, Hokkaido University, Sapporo-shi, Hokkaido, Japan
| | - Seiichi Kitahama
- Department of Metabolic and Bariatric Surgery, Center for Obesity, Diabetes and Endocrinology, Chibune General Hospital, Osaka-shi, Osaka, Japan
| | - Masako Ueyama
- Sleep Apnea Syndrome Treatment Center, Fukujuji Hospital, Japan Anti-Tuberculosis Association, Kiyose-shi, Tokyo, Japan
| | - Takumi Murakami
- Advanced Genomics Center, National Institute of Genetics, Yata, Mishima-shi, Shizuoka, Japan
- School of Life Science and Technology, Institute of Science Tokyo, Meguro-ku, Tokyo, Japan
| | - Shinsuke Inuki
- Department of Bioorganic Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Junichiro Irie
- Department of Endocrinology, Metabolism and Nephrology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Noriko Satoh-Asahara
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, Kyoto, Japan
| | - Hirokazu Toju
- Laboratory of Ecosystems and Coevolution, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Center for Living Systems Information Science (CeLiSIS), Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hiroshi Mori
- Advanced Genomics Center, National Institute of Genetics, Yata, Mishima-shi, Shizuoka, Japan
| | - Shinji Nakaoka
- Laboratory of Mathematical Biology, Faculty of Advanced Life Science, Hokkaido University, Sapporo-shi, Hokkaido, Japan
| | - Tomoya Yamashita
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe-shi, Hyogo, Japan
| | - Atsushi Toyoda
- Advanced Genomics Center, National Institute of Genetics, Yata, Mishima-shi, Shizuoka, Japan
| | - Kenji Yamamoto
- Center for Innovative and Joint Research, Wakayama University, Wakayama-shi, Wakayama, Japan
| | - Hiroaki Ohno
- Department of Bioorganic Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan
| | - Takane Katayama
- Laboratory of Molecular Biology and Bioresponse, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Hiroshi Itoh
- Department of Endocrinology, Metabolism and Nephrology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan
| | - Ikuo Kimura
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, Japan.
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan.
- Department of Molecular Endocrinology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan.
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan.
- Department of Moonshot Research and Development Program, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan.
| |
Collapse
|
28
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
29
|
Li Y, Gao R, Yang Z, Zong H, Li Y. Liraglutide modulates lipid metabolism via ZBTB20-LPL pathway. Life Sci 2025; 360:123267. [PMID: 39608448 DOI: 10.1016/j.lfs.2024.123267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE To investigate the mechanism of liraglutide affecting lipid metabolism by regulating lipolysis and lipogenesis in cells and ob/ob mice. METHODS 3 T3-L1 cells were treated with liraglutide in vitro, and differentially expressed genes were screened by RNA sequencing. Gene Ontology (GO) and KEGG (Kvoto Encyclopedia of Genes and Genomes) enrichment analyses identified target genes for lipid regulation of liraglutide. 3 T3-L1 preadipocytes were induced to differentiate into adipocytes using a "cocktail method". Western blot and immunofluorescence were used to detect the expression of target genes and the lipid regulatory effect of liraglutide. 3 T3-L1 preadipocytes were transfected with lentivirus overexpressing Zbtb20 to study its role in adipogenesis, and gene expression was analyzed by RT-qPCR and Western blot. In vivo, ob/ob mice were subcutaneously injected with liraglutide or saline for 4 weeks. Blood lipids, adipose tissue volume and adipocyte size were detected. Immunohistochemical analysis and RT-qPCR were used to detect the expression of target genes in adipose tissue. RESULTS Liraglutide reduced lipid droplets and TG levels and altered the expression of genes related to fatty acid metabolism, lipogenesis, fatty acid oxidation, and adipocyte browning. The results of PCR, Western blot and immunofluorescence confirmed that liraglutide could regulate the adipogenesis by downregulating the transcriptional suppressor ZBTB20, and overexpression of Zbtb20 inhibited the expression of LPL, the key enzyme for lipohydrolysis. CONCLUSIONS Liraglutide regulates lipid metabolism through ZBTB20-LPL pathway to reveal its molecular mechanism.
Collapse
Affiliation(s)
- Yue Li
- Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan- Hospital, Jinan, Shandong, China; Department of Medicine, Qilu Institute of Technology, Jinan, China.
| | - Rui Gao
- Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan- Hospital, Jinan, Shandong, China.
| | - Zhiyan Yang
- Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan- Hospital, Jinan, Shandong, China; Department of Pharmacy, Jining No.1 People's Hospital, Jining, China.
| | - Huiying Zong
- Department of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Yan Li
- Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan- Hospital, Jinan, Shandong, China.
| |
Collapse
|
30
|
Harada K, Wada E, Osuga Y, Shimizu K, Uenoyama R, Hirai MY, Maekawa F, Miyazaki M, Hayashi YK, Nakamura K, Tsuboi T. Intestinal butyric acid-mediated disruption of gut hormone secretion and lipid metabolism in vasopressin receptor-deficient mice. Mol Metab 2025; 91:102072. [PMID: 39668067 PMCID: PMC11728074 DOI: 10.1016/j.molmet.2024.102072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024] Open
Abstract
OBJECTIVES 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. METHODS 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-/- mice), V1bR-deficient mice (V1bR-/- mice), and V1aR/V1bR-double deficient mice (V1aR-/-V1bR-/-mice). RESULTS AVP induced the increase in intracellular Ca2+ levels and GLP-1 secretion from GLUTag cells in a V1aR and V1bR-dependent manner. AVP receptor-deficient mice, particularly V1aR-/-V1bR-/- mice, demonstrated impaired secretion of GLP-1 and peptide YY secreted by enteroendocrine L cells. V1aR-/-V1bR-/-mice also exhibited abnormal lipid accumulation in the brown adipose tissue and skeletal muscle. We discovered that V1aR-/-V1bR-/- 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. CONCLUSIONS 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.
Collapse
Affiliation(s)
- Kazuki Harada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo 153-8902, Japan; Health and Environmental Risk Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki 305-8506, Japan
| | - Eiji Wada
- Department of Pathophysiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku, Tokyo 160-8402, Japan
| | - Yuri Osuga
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo 153-8902, Japan
| | - Kie Shimizu
- Department of Pharmacology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan; Division of Life Sciences, Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura, Saitama 338-8570, Japan
| | - Reiko Uenoyama
- The United Graduate School of Agricultural Sciences, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Masami Yokota Hirai
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama-city, Kanagawa 230-0045, Japan
| | - Fumihiko Maekawa
- Health and Environmental Risk Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki 305-8506, Japan
| | - Masao Miyazaki
- The United Graduate School of Agricultural Sciences, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Yukiko K Hayashi
- Department of Pathophysiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku, Tokyo 160-8402, Japan
| | - Kazuaki Nakamura
- Department of Pharmacology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan; Division of Life Sciences, Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura, Saitama 338-8570, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo 153-8902, Japan.
| |
Collapse
|
31
|
Kononova YA, Tuchina TP, Babenko AY. Brown and Beige Adipose Tissue: One or Different Targets for Treatment of Obesity and Obesity-Related Metabolic Disorders? Int J Mol Sci 2024; 25:13295. [PMID: 39769065 PMCID: PMC11677471 DOI: 10.3390/ijms252413295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
The failure of the fight against obesity makes us turn to new goals in its treatment. Now, brown adipose tissue has attracted attention as a promising target for the treatment of obesity and associated metabolic disorders such as insulin resistance, dyslipidemia, and glucose tolerance disorders. Meanwhile, the expansion of our knowledge has led to awareness about two rather different subtypes: classic brown and beige (inducible brown) adipose tissue. These subtypes have different origin, differences in the expression of individual genes but also a lot in common. Both tissues are thermogenic, which means that, by increasing energy consumption, they can improve their balance with excess intake. Both tissues are activated in response to specific inducers (cold, beta-adrenergic receptor activation, certain food and drugs), but beige adipose tissue transdifferentiates back into white adipose tissue after the cessation of inducing action, while classic brown adipose tissue persists, but its activity decreases. In this review, we attempted to understand whether there are differences in the effects of different groups of thermogenesis-affecting drugs on these tissues. The analysis showed that this area of research is rather sparse and requires close attention in further studies.
Collapse
Affiliation(s)
- Yulia A. Kononova
- World-Class Scientific Center “Center for Personalized Medicine”, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Taisiia P. Tuchina
- Endocrinology Department, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Alina Yu. Babenko
- World-Class Scientific Center “Center for Personalized Medicine”, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| |
Collapse
|
32
|
Díaz-Castro F, Morselli E, Claret M. Interplay between the brain and adipose tissue: a metabolic conversation. EMBO Rep 2024; 25:5277-5293. [PMID: 39558137 PMCID: PMC11624209 DOI: 10.1038/s44319-024-00321-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
The central nervous system and adipose tissue interact through complex communication. This bidirectional signaling regulates metabolic functions. The hypothalamus, a key homeostatic brain region, integrates exteroceptive and interoceptive signals to control appetite, energy expenditure, glucose, and lipid metabolism. This regulation is partly achieved via the nervous modulation of white (WAT) and brown (BAT) adipose tissue. In this review, we highlight the roles of sympathetic and parasympathetic innervation in regulating WAT and BAT activities, such as lipolysis and thermogenesis. Adipose tissue, in turn, plays a dual role as an energy reservoir and an endocrine organ, secreting hormones that influence brain function and metabolic health. In addition, this review focuses on recently uncovered communication pathways, including extracellular vesicles and neuro-mesenchymal units, which add new layers of regulation and complexity to the brain-adipose tissue interaction. Finally, we also examine the consequences of disrupted communication between the brain and adipose tissue in metabolic disorders like obesity and type-2 diabetes, emphasizing the potential for new therapeutic strategies targeting these pathways to improve metabolic health.
Collapse
Affiliation(s)
- Francisco Díaz-Castro
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile
- Physiology Department, Biological Science Faculty, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Faculty of Medicine and Sciences, Department of Basic Sciences, Universidad San Sebastián, Santiago de Chile, Chile.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- IBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
33
|
Rossetti CL, Andrade IS, Fonte Boa LF, Neves MB, Fassarella LB, Bertasso IM, Souza MDGCD, Bouskela E, Lisboa PC, Takyia CM, Trevenzoli IH, Fortunato RS, Carvalho DPD. Liraglutide prevents body and fat mass gain in ovariectomized Wistar rats. Mol Cell Endocrinol 2024; 594:112374. [PMID: 39306226 DOI: 10.1016/j.mce.2024.112374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Estrogens exert beneficial metabolic effects by reducing food intake and enhancing energy expenditure through both central and peripheral mechanisms. The decrease of estrogen, as occurs in ovariectomy (OVX), leads to metabolic disturbances, such as increased body weight, adipose tissue mass, basal blood glucose, and impaired glucose tolerance. These effects can be reversed by reintroducing estrogen. GLP-1 and its receptor agonists, known for their antihyperglycemic properties, also exhibit anorexigenic effects. Besides that, research indicates that GLP-1 analogs can induce metabolic changes peripherally, such as increased fatty acid oxidation and inhibited lipogenesis. Given the shared metabolic actions of GLP-1 and estrogens, we explored whether liraglutide, a GLP-1 agonist, could mitigate the metabolic effects of estrogen deficiency. We tested this hypothesis using ovariectomized rats, a model that simulates menopausal estrogen deficiency, and treated them with either liraglutide or 17β-Estradiol benzoate for 21 days. Ovariectomy resulted in elevated DPP-IV activity in both plasma and inguinal white adipose tissue (iWAT). While estrogen replacement effectively countered the DPP-IV increase in both plasma and iWAT, liraglutide only prevented the rise in iWAT DPP-IV activity. Liraglutide prevented body weight and fat mass gain after ovariectomy to the same extent as estradiol treatment. This can be explained by the lower food intake and food efficiency caused by estradiol and liraglutide. However, liraglutide was associated with increased pro-inflammatory cytokines and inflammatory cells in white adipose tissue. Further research is crucial to fully understand the potential benefits and risks of using GLP-1 receptor agonists in the context of menopause.
Collapse
Affiliation(s)
- Camila Lüdke Rossetti
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil; Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, USA
| | - Iris Soares Andrade
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Fernando Fonte Boa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Barbosa Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa Brito Fassarella
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iala Milene Bertasso
- Laboratorio de Fisiologia Endócrina, Instituto de Biologia, Universidade Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria das Graças Coelho de Souza
- Laboratório de Pesquisa Clínica e Experimental em Biologia Vascular (BioVasc), Universidade Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliete Bouskela
- Laboratório de Pesquisa Clínica e Experimental em Biologia Vascular (BioVasc), Universidade Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Cristina Lisboa
- Laboratorio de Fisiologia Endócrina, Instituto de Biologia, Universidade Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christina Maeda Takyia
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Ciências Cirúrgicas, Faculdade de Medicina, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isis Hara Trevenzoli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo Soares Fortunato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Denise Pires de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Model JFA, Normann RS, Vogt ÉL, Dentz MV, de Amaral M, Xu R, Bachvaroff T, Spritzer PM, Chung JS, Vinagre AS. Interactions between glucagon like peptide 1 (GLP-1) and estrogens regulates lipid metabolism. Biochem Pharmacol 2024; 230:116623. [PMID: 39542180 DOI: 10.1016/j.bcp.2024.116623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Obesity, characterized by excessive fat accumulation in white adipose tissue (WAT), is linked to numerous health issues, including insulin resistance (IR), and type 2 diabetes mellitus (DM2). The distribution of adipose tissue differs by sex, with men typically exhibiting android adiposity and pre-menopausal women displaying gynecoid adiposity. After menopause, women have an increased risk of developing android-type obesity, IR, and DM2. Glucagon-like peptide 1 (GLP-1) receptor agonists (GLP-1RAs) are important in treating obesity and DM2 by regulating insulin secretion, impacting glucose and lipid metabolism. GLP-1Rs are found in various tissues including the pancreas, brain, and adipose tissue. Studies suggest GLP-1RAs and estrogen replacement therapies have similar effects on tissues like the liver, central nervous system, and WAT, probably by converging pathways involving protein kinases. To investigate these interactions, female rats underwent ovariectomy (OVR) to promote a state of estrogen deficiency. After 20 days, the rats were euthanized and the tissues were incubated with 10 μM of liraglutide, a GLP-1RA. Results showed significant changes in metabolic parameters: OVR increased lipid catabolism in perirenal WAT and basal lipolysis in subcutaneous WAT, while liraglutide treatment enhanced stimulated lipolysis in subcutaneous WAT. Liver responses included increased stimulated lipolysis with liraglutide. Transcriptome analysis revealed distinct gene expression patterns in WAT of OVR rats and those treated with GLP-1RA, highlighting pathways related to lipid and glucose metabolism. Functional enrichment analysis showed estrogen's pivotal role in these pathways, influencing genes involved in lipid metabolism regulation. Overall, the study underscores GLP-1RA acting directly on adipose tissues and highlights the complex interactions between GLP-1 and estrogen in regulating metabolism, suggesting potential synergistic therapeutic effects in treating metabolic disorders like obesity and DM2.
Collapse
Affiliation(s)
- Jorge F A Model
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Rafaella S Normann
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Éverton L Vogt
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Maiza Von Dentz
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Marjoriane de Amaral
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Rui Xu
- Institute of Marine and Environmental Technology (IMET), University of Maryland Center for Environmental Science, Baltimore, MD 21202, USA
| | - Tsvetan Bachvaroff
- Institute of Marine and Environmental Technology (IMET), University of Maryland Center for Environmental Science, Baltimore, MD 21202, USA
| | - Poli Mara Spritzer
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - J Sook Chung
- Institute of Marine and Environmental Technology (IMET), University of Maryland Center for Environmental Science, Baltimore, MD 21202, USA
| | - Anapaula S Vinagre
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
35
|
Thakur P, Baraskar K, Shrivastava VK, Medhi B. Cross-talk between adipose tissue and microbiota-gut-brain-axis in brain development and neurological disorder. Brain Res 2024; 1844:149176. [PMID: 39182900 DOI: 10.1016/j.brainres.2024.149176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/25/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
The gut microbiota is an important factor responsible for the physiological processes as well as pathogenesis of host. The communication between central nervous system (CNS) and microbiota occurs by different pathways i.e., chemical, neural, immune, and endocrine. Alteration in gut microbiota i.e., gut dysbiosis causes alteration in the bidirectional communication between CNS and gut microbiota and linked to the pathogenesis of neurological and neurodevelopmental disorder. Therefore, now-a-days microbiota-gut-brain-axis (MGBA) has emerged as therapeutic target for the treatment of metabolic disorder. But, experimental data available on MGBA from basic research has limited application in clinical study. In present study we first summarized molecular mechanism of microbiota interaction with brain physiology and pathogenesis via collecting data from different sources i.e., PubMed, Scopus, Web of Science. Furthermore, evidence shows that adipose tissue (AT) is active during metabolic activities and may also interact with MGBA. Hence, in present study we have focused on the relationship among MGBA, brown adipose tissue, and white adipose tissue. Along with this, we have also studied functional specificity of AT, and understanding heterogeneity among MGBA and different types of AT. Therefore, molecular interaction among them may provide therapeutic target for the treatment of neurological disorder.
Collapse
Affiliation(s)
- Pratibha Thakur
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India.
| | - Kirti Baraskar
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Vinoy K Shrivastava
- Endocrinology Unit, Bioscience Department, Barkatullah University, Bhopal, Madhya Pradesh 462026, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab 160012, India.
| |
Collapse
|
36
|
Saglietto A, Falasconi G, Penela D, Francia P, Sau A, Ng FS, Dusi V, Castagno D, Gaita F, Berruezo A, De Ferrari GM, Anselmino M. Glucagon-like peptide-1 receptor agonist semaglutide reduces atrial fibrillation incidence: A systematic review and meta-analysis. Eur J Clin Invest 2024; 54:e14292. [PMID: 39058274 DOI: 10.1111/eci.14292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are new anti-hyperglycaemic drugs with proven cardiovascular (CV) benefit in diabetic and non-diabetic patients at high CV risk. Despite a neutral class effect on arrhythmia risk, data on semaglutide suggest a possible drug-specific benefit in reducing atrial fibrillation (AF) occurrence. OBJECTIVE To perform a meta-analysis of randomized clinical trials (RCTs) to assess the risk of incident AF in patients treated with semaglutide compared to placebo. METHODS AND RESULTS Ten RCTs were included in the analysis. Study population encompassed 12,651 patients (7285 in semaglutide and 5366 in placebo arms), with median follow-up of 68 months. A random effect meta-analytic model was adopted to pool relative risk (RR) of incident AF. Semaglutide reduces the risk of AF by 42% (RR .58, 95% CI .40-.85), with low heterogeneity across the studies (I2 0%). At subgroup analysis, no differences emerged between oral and subcutaneous administration (oral: RR .53, 95% CI .23-1.24, I2 0%; subcutaneous: RR .59, 95% CI .39-.91, I2 0%; p-value .83). In addition, meta-regression analyses did not show any potential influence of baseline study covariates, in particular the proportion of diabetic patients (p-value .14) and body mass index (BMI) (p-value .60). CONCLUSIONS Semaglutide significantly reduces the occurrence of incident AF by 42% as compared to placebo in individuals at high CV risk, mainly affected by type 2 diabetes mellitus. This effect appears to be consistent independently of the route of administration of the drug (oral or subcutaneous), the presence of underlying diabetes and BMI.
Collapse
Affiliation(s)
- Andrea Saglietto
- Division of Cardiology, Cardiovascular and Thoracic Department, "Citta della Salute e della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
- Heart Institute, Teknon Medical Centre, Barcelona, Spain
| | - Giulio Falasconi
- Heart Institute, Teknon Medical Centre, Barcelona, Spain
- Campus Clínic, University of Barcelona, Barcelona, Spain
- Arrhythmology Department, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Diego Penela
- Heart Institute, Teknon Medical Centre, Barcelona, Spain
- Arrhythmology Department, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Pietro Francia
- Heart Institute, Teknon Medical Centre, Barcelona, Spain
- Division of Cardiology, Department of Clinical and Molecular Medicine, St. Andrea Hospital, Sapienza University
| | - Arunashis Sau
- National Heart and Lung Institute, Hammersmith Campus, Imperial College London, London, UK
- Department of Cardiology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Fu Siong Ng
- National Heart and Lung Institute, Hammersmith Campus, Imperial College London, London, UK
- Department of Cardiology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
- Department of Cardiology, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Veronica Dusi
- Division of Cardiology, Cardiovascular and Thoracic Department, "Citta della Salute e della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Davide Castagno
- Division of Cardiology, Cardiovascular and Thoracic Department, "Citta della Salute e della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fiorenzo Gaita
- Department of Medical Sciences, University of Turin, Turin, Italy
- Cardiology Unit, J Medical, Turin, Italy
| | | | - Gaetano Maria De Ferrari
- Division of Cardiology, Cardiovascular and Thoracic Department, "Citta della Salute e della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Matteo Anselmino
- Division of Cardiology, Cardiovascular and Thoracic Department, "Citta della Salute e della Scienza" Hospital, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
37
|
Araj-Khodaei M, Ayati MH, Azizi Zeinalhajlou A, Novinbahador T, Yousefi M, Shiri M, Mahmoodpoor A, Shamekh A, Namazi N, Sanaie S. Berberine-induced glucagon-like peptide-1 and its mechanism for controlling type 2 diabetes mellitus: a comprehensive pathway review. Arch Physiol Biochem 2024; 130:678-685. [PMID: 37921026 DOI: 10.1080/13813455.2023.2258559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/05/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION A growing number of studies have thus far showed the association between type 2 diabetes mellitus (DM) and the intestinal microbiome homoeostasis. As reported, the gut microflora can be significantly different in patients with type 2 DM (T2DM) compared to those in healthy individuals. METHODS The authors collected the relevant articles published until 2022 and these are carefully selected from three scientific databases based on keywords. DISCUSSION This review highlights research on the anti-diabetic properties of berberine (BBR)-induced glucagon-like peptide-1 (GLP-1), as a glucose-lowering factor and a balance regulator in the microbial flora of the intestines, which plays an important role in adjusting the signalling pathways affecting insulin secretion. RESULTS Considering the anti-diabetic characteristics of the BBR-induced GLP-1, BBR makes a promising complementary treatment for reducing the clinical symptoms of DM by reducing the hyperglycaemia. Berberin might be a safe and effective drug for T2DM with little or no adverse effects.
Collapse
Affiliation(s)
| | - Mohammad Hossein Ayati
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Azizi Zeinalhajlou
- Department of Geriatric Health, Faculty of Health Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tannaz Novinbahador
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Shiri
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Student Research Committee, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazli Namazi
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sarvin Sanaie
- Research Center of Psychiatry and Behavioral Sciences, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
38
|
López M, Gualillo O. Rheumatic diseases and metabolism: where centre and periphery meet. Nat Rev Rheumatol 2024; 20:783-794. [PMID: 39478099 DOI: 10.1038/s41584-024-01178-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 11/26/2024]
Abstract
Over the past few decades, the connection between metabolism and various inflammatory and rheumatic diseases has been an area of active investigation. Nonetheless, the precise mechanisms underlying these relationships remain a topic of ongoing debate, owing in part to conflicting data. This discrepancy can be attributed to the predominant focus on peripheral mechanisms in research into the metabolic consequences of rheumatic diseases. However, a wealth of evidence supports the notion that the central nervous system, specifically the hypothalamus, has an important influence on metabolic homeostasis. Notably, links have been established between crucial hypothalamic mechanisms responsible for regulating energy balance (including food intake, thermogenesis, and glucose and lipid metabolism), such as AMP-activated protein kinase, and the pathophysiology of rheumatoid arthritis. This Review aims to comprehensively examine the current understanding of central metabolic control in rheumatic diseases and explore potential therapeutic options that target this pathophysiological mechanism.
Collapse
Affiliation(s)
- Miguel López
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, Spain.
| | - Oreste Gualillo
- Servizo Galego de Saude (SERGAS)-Instituto de Investigación Sanitaria de Santiago (IDIS), the Neuroendocrine Interactions in Rheumatology and Inflammatory Disease (NEIRID) Lab, Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| |
Collapse
|
39
|
Dongdem JT, Etornam AE, Beletaa S, Alidu I, Kotey H, Wezena CA. The β 3-Adrenergic Receptor: Structure, Physiopathology of Disease, and Emerging Therapeutic Potential. Adv Pharmacol Pharm Sci 2024; 2024:2005589. [PMID: 39640497 PMCID: PMC11620816 DOI: 10.1155/2024/2005589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
The discovery and characterization of the signal cascades of the β-adrenergic receptors have made it possible to effectively target the receptors for drug development. β-Adrenergic receptors are a class A rhodopsin type of G protein-coupled receptors (GPCRs) that are stimulated mainly by catecholamines and therefore mediate diverse effects of the parasympathetic nervous system in eliciting "fight or flight" type responses. They are detectable in several human tissues where they control a plethora of physiological processes and therefore contribute to the pathogenesis of several disease conditions. Given the relevance of the β-adrenergic receptor as a molecular target for many pathological conditions, this comprehensive review aims at providing an in-depth exploration of the recent advancements in β3-adrenergic receptor research. More importantly, we delve into the prospects of the β3-adrenergic receptor as a therapeutic target across a variety of clinical domains.
Collapse
Affiliation(s)
- Julius T. Dongdem
- Department of Chemical Pathology, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Axandrah E. Etornam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Solomon Beletaa
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Issah Alidu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Hassan Kotey
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Cletus A. Wezena
- Department of Microbiology, Faculty of Biosciences, University for Development Studies, Tamale, Northern Region, Ghana
| |
Collapse
|
40
|
Dimitri P, Roth CL. Treatment of Hypothalamic Obesity With GLP-1 Analogs. J Endocr Soc 2024; 9:bvae200. [PMID: 39703362 PMCID: PMC11655849 DOI: 10.1210/jendso/bvae200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Congenital and acquired damage to hypothalamic nuclei or neuronal circuits controlling satiety and energy expenditure results in hypothalamic obesity (HO). To date, successful weight loss and satiety has only been achieved in a limited number of affected patients across multiple drug trials. Glucagon-like peptide-1 (GLP-1) acts via central pathways that are independent from the hypothalamus to induce satiety. GLP-1 receptor agonists (GLP-1RAs) may provide an alternative approach to treating HO. Methods We performed a comprehensive search in Medline, Google Scholar, and clinical trials registries (ClinicalTrials.gov; clinicaltrialsregister.eur). This nonsystematic literature review was conducted to identify scientific papers published from January 2005 to February 2024 using the Pubmed and Embase databases. Key words used were GLP-1, GLP-1RA, hypothalamic obesity, suprasellar tumor, and craniopharyngioma. Results Our search identified 7 case studies, 5 case series, and 2 published clinical trials relating to the use of GLP-1RAs in HO. All case studies demonstrated weight loss and improved metabolic function. In contrast, results from case series were variable, with some showing no weight loss and others demonstrating moderate to significant weight loss and improved metabolic parameters. In the ECHO clinical trial, nearly half the subjects randomized to weekly exenatide showed reduced body mass index (BMI). Paradoxically, BMI reduction was greater in patients with more extensive hypothalamic injuries. Conclusion GLP-1RAs potentially offer a new approach to treating HO. There is a need to stratify patients who are more likely to respond. Further randomized controlled trials are required to determine their efficacy either in isolation or combined with other therapies.
Collapse
Affiliation(s)
- Paul Dimitri
- The Department of Paediatric Endocrinology, Sheffield Children's NHS Foundation Trust, Sheffield, S10 2TH, UK
- University of Sheffield, Sheffield, S10 2TN, UK
| | - Christian L Roth
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
41
|
Aguggia J, Fernandez G, Cassano D, Mustafá ER, Rodríguez SS, Cantel S, Fehrentz JA, Raingo J, Schiöth HB, Habib AM, De Francesco PN, Perello M. Selective Colocalization of GHSR and GLP-1R in a Subset of Hypothalamic Neurons and Their Functional Interaction. Endocrinology 2024; 166:bqae160. [PMID: 39737802 DOI: 10.1210/endocr/bqae160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Indexed: 01/01/2025]
Abstract
The GH secretagogue receptor (GHSR) and the glucagon-like peptide-1 receptor (GLP-1R) are G protein-coupled receptors with critical, yet opposite, roles in regulating energy balance. Interestingly, these receptors are expressed in overlapping brain regions. However, the extent to which they target the same neurons and engage in molecular crosstalk remains unclear. To explore the potential colocalization of GHSR and GLP-1R in specific neurons, we performed detailed mapping of cells positive for both receptors using GHSR-eGFP reporter mice or wild-type mice infused with fluorescent ghrelin, alongside an anti-GLP-1R antibody. We found that GHSR+ and GLP-1R+ cells are largely segregated in the mouse brain. The highest overlap was observed in the hypothalamic arcuate nucleus, where 15% to 20% of GHSR+ cells were also GLP-1R+ cells. Additionally, we examined RNA-sequencing datasets from mouse and human brains to assess the fraction and distribution of neurons expressing both receptors, finding that double-positive Ghsr+/Glp1r+ cells are highly segregated, with a small subset of double-positive Ghsr+/Glp1r+ cells representing <10% of all Ghsr+ or Glp1r+ cells, primarily enriched in the hypothalamus. Furthermore, we conducted functional studies using patch-clamp recordings in a heterologous expression system to assess potential crosstalk in regulating presynaptic calcium channels. We provide the first evidence that liraglutide-evoked GLP-1R activity inhibits presynaptic channels, and that the presence of one GPCR attenuates the inhibitory effects of ligand-evoked activity mediated by the other on presynaptic calcium channels. In conclusion, while GHSR and GLP-1R can engage in molecular crosstalk, they are largely segregated across most neuronal types within the brain.
Collapse
Affiliation(s)
- Julieta Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Emilio R Mustafá
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Silvia S Rodríguez
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Jesica Raingo
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, 751 24 Uppsala, Sweden
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, 751 24 Uppsala, Sweden
| |
Collapse
|
42
|
Quagliariello V, Canale ML, Bisceglia I, Iovine M, Giordano V, Giacobbe I, Scherillo M, Gabrielli D, Maurea C, Barbato M, Inno A, Berretta M, Tedeschi A, Oliva S, Greco A, Maurea N. Glucagon-like Peptide 1 Receptor Agonists in Cardio-Oncology: Pathophysiology of Cardiometabolic Outcomes in Cancer Patients. Int J Mol Sci 2024; 25:11299. [PMID: 39457081 PMCID: PMC11508560 DOI: 10.3390/ijms252011299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Cancer patients, especially long cancer survivors, are exposed to several cardio-metabolic diseases, including diabetes, heart failure, and atherosclerosis, which increase their risk of cardiovascular mortality. Therapy with glucagon-like peptide 1 (GLP1) receptor agonists demonstrated several beneficial cardiovascular effects, including atherosclerosis and heart failure prevention. Cardiovascular outcome trials (CVOTs) suggest that GLP-1 RA could exert cardiorenal benefits and systemic anti-inflammatory effects in patients with type-2 diabetes through the activation of cAMP and PI3K/AkT pathways and the inhibition of NLRP-3 and MyD88. In this narrative review, we highlight the biochemical properties of GLP-1 RA through a deep analysis of the clinical and preclinical evidence of the primary prevention of cardiomyopathies. The overall picture of this review encourages the study of GLP-1 RA in cancer patients with type-2 diabetes, as a potential primary prevention strategy against heart failure and atherosclerosis.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | | | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy;
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Vienna Giordano
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Marino Scherillo
- Division of Cardiology, Hospital San Pio Benevento (BN), 82100 Benevento, Italy;
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy;
| | - Carlo Maurea
- Department of Medicine, University of Salerno, 84084 Fisciano, Italy;
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Andrea Tedeschi
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
| | - Stefano Oliva
- UOSD Cardiologia di Interesse Oncologico IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Alessandra Greco
- Divisione di Cardiologia, Fondazione IRCCS San Matteo Hospital, Viale Golgi 19, 27100 Pavia, Italy;
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| |
Collapse
|
43
|
Xu Z, Li H, Cao G, Li P, Zhou H, Sun Y. The protective role of brown adipose tissue in cardiac cell damage after myocardial infarction and heart failure. Lipids Health Dis 2024; 23:338. [PMID: 39415186 PMCID: PMC11481725 DOI: 10.1186/s12944-024-02326-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/06/2024] [Indexed: 10/18/2024] Open
Abstract
Acute myocardial infarction (AMI) and related cardiovascular disease complications are the leading causes of mortality worldwide. Brown adipose tissue (BAT) is thermogenic and characterized by the uncoupling protein expression. Recent studies have found that in cardiovascular diseases, activated BAT can effectively improve the prognosis of AMI and concurrent heart failure through intercellular communication. However, a clear and systematic understanding of the myocardial protective mechanism of BAT after AMI is lacking, especially in the endocrine function of BAT. This review describes the effects of BAT on various cells in the heart after AMI. BAT plays a protective role on cardiac cells and fibroblasts during ischemia/reperfusion (I/R), myocardial remodeling, and myocardial fibrosis. This review also discusses the changes caused by BAT activation in different stages of heart failure. Finally, this review summarizes the treatment methods that target BAT to improve AMI. Further in-depth researches are still needed to clarify the underlying mechanism of the connection between BAT and different cells in cardiac tissue in order to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Zhe Xu
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hong Li
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guojie Cao
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Panpan Li
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haitao Zhou
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Sun
- Department of Geriatric Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
44
|
Wang F, Huynh PM, An YA. Mitochondrial Function and Dysfunction in White Adipocytes and Therapeutic Implications. Compr Physiol 2024; 14:5581-5640. [PMID: 39382163 DOI: 10.1002/cphy.c230009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
45
|
Yu J, Gao M, Wang L, Guo X, Liu X, Sheng M, Cheng S, Guo Y, Wang J, Zhao C, Guo W, Zhang Z, Liu Y, Hu C, Ma X, Xie C, Zhang Q, Xu L. An insoluble cellulose nanofiber with robust expansion capacity protects against obesity. Int J Biol Macromol 2024; 277:134401. [PMID: 39097049 DOI: 10.1016/j.ijbiomac.2024.134401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
An imbalance between energy intake and energy expenditure predisposes obesity and its related metabolic diseases. Soluble dietary fiber has been shown to improve metabolic homeostasis mainly via microbiota reshaping. However, the application and metabolic effects of insoluble fiber are less understood. Herein, we employed nanotechnology to design citric acid-crosslinked carboxymethyl cellulose nanofibers (CL-CNF) with a robust capacity of expansion upon swelling. Supplementation with CL-CNF reduced food intake and delayed digestion rate in mice by occupying stomach. Besides, CL-CNF treatment mitigated diet-induced obesity and insulin resistance in mice with enhanced energy expenditure, as well as ameliorated inflammation in adipose tissue, intestine and liver and reduced hepatic steatosis, without any discernible signs of toxicity. Additionally, CL-CNF supplementation resulted in enrichment of probiotics such as Bifidobacterium and decreased in the relative abundances of deleterious microbiota expressing bile salt hydrolase, which led to increased levels of conjugated bile acids and inhibited intestinal FXR signaling to stimulate the release of GLP-1. Taken together, our findings demonstrate that CL-CNF administration protects mice from diet-induced obesity and metabolic dysfunction by reducing food intake, enhancing energy expenditure and remodeling gut microbiota, making it a potential therapeutic strategy against metabolic diseases.
Collapse
Affiliation(s)
- Jian Yu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingyuan Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaodi Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Maozheng Sheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shimiao Cheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yingying Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiawen Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Cheng Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wenxiu Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhe Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yameng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xinran Ma
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China; Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, China.
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Qiang Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
46
|
Chen B, Yu X, Horvath-Diano C, Ortuño MJ, Tschöp MH, Jastreboff AM, Schneeberger M. GLP-1 programs the neurovascular landscape. Cell Metab 2024; 36:2173-2189. [PMID: 39357509 DOI: 10.1016/j.cmet.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Readily available nutrient-rich foods exploit our inherent drive to overconsume, creating an environment of overnutrition. This transformative setting has led to persistent health issues, such as obesity and metabolic syndrome. The development of glucagon-like peptide-1 receptor (GLP-1R) agonists reveals our ability to pharmacologically manage weight and address metabolic conditions. Obesity is directly linked to chronic low-grade inflammation, connecting our metabolic environment to neurodegenerative diseases. GLP-1R agonism in curbing obesity, achieved by impacting appetite and addressing associated metabolic defects, is revealing additional benefits extending beyond weight loss. Whether GLP-1R agonism directly impacts brain health or does so indirectly through improved metabolic health remains to be elucidated. In exploring the intricate connection between obesity and neurological conditions, recent literature suggests that GLP-1R agonism may have the capacity to shape the neurovascular landscape. Thus, GLP-1R agonism emerges as a promising strategy for addressing the complex interplay between metabolic health and cognitive well-being.
Collapse
Affiliation(s)
- Bandy Chen
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Xiaofei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Claudia Horvath-Diano
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - María José Ortuño
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Matthias H Tschöp
- Helmholtz Zentrum München, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany
| | - Ania M Jastreboff
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - Marc Schneeberger
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA; Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
47
|
Petersen EA, Blom I, Melander SA, Al-Rubai M, Vidotto M, Dalgaard LT, Karsdal MA, Henriksen K, Larsen S, Larsen AT. DACRA induces profound weight loss, satiety control, and increased mitochondrial respiratory capacity in adipose tissue. Int J Obes (Lond) 2024; 48:1421-1429. [PMID: 38879729 DOI: 10.1038/s41366-024-01564-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND AND OBJECTIVES Dual amylin and calcitonin receptor agonists (DACRAs) are therapeutic candidates in the treatment of obesity with beneficial effects on weight loss superior to suppression of food intake. Hence, suggesting effects on energy expenditure by possibly targeting mitochondria in metabolically active tissue. METHODS Male rats with HFD-induced obesity received a DACRA, KBP-336, every third day for 8 weeks. Upon study end, mitochondrial respiratory capacity (MRC), - enzyme activity, - transcriptional factors, and -content were measured in perirenal (pAT) and inguinal adipose tissue. A pair-fed group was included to examine food intake-independent effects of KBP-336. RESULTS A vehicle-corrected weight loss (23.4 ± 2.8%) was achieved with KBP-336, which was not observed to the same extent with the food-restricted weight loss (12.4 ± 2.8%) (P < 0.001). Maximal coupled respiration supported by carbohydrate and lipid-linked substrates was increased after KBP-336 treatment independent of food intake in pAT (P < 0.01). Moreover, oligomycin-induced leak respiration and the activity of citrate synthase and β-hydroxyacetyl-CoA-dehydrogenase were increased with KBP-336 treatment (P < 0.05). These effects occurred without changes in mitochondrial content in pAT. CONCLUSIONS These findings demonstrate favorable effects of KBP-336 on MRC in adipose tissue, indicating an increased energy expenditure and capacity to utilize fatty acids. Thus, providing more mechanistic insight into the DACRA-induced weight loss.
Collapse
Affiliation(s)
- Emilie A Petersen
- Nordic Bioscience, Herlev, Denmark.
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Ida Blom
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Mays Al-Rubai
- Nordic Bioscience, Herlev, Denmark
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | | | - Louise T Dalgaard
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Morten A Karsdal
- Nordic Bioscience, Herlev, Denmark
- KeyBioscience AG, Stans, Switzerland
| | - Kim Henriksen
- Nordic Bioscience, Herlev, Denmark
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- KeyBioscience AG, Stans, Switzerland
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | | |
Collapse
|
48
|
Ji L, He X, Min X, Yang H, Wu W, Xu H, Chen J, Mei A. Glucagon-like peptide-1 receptor agonists in neoplastic diseases. Front Endocrinol (Lausanne) 2024; 15:1465881. [PMID: 39371922 PMCID: PMC11449759 DOI: 10.3389/fendo.2024.1465881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/28/2024] [Indexed: 10/08/2024] Open
Abstract
Glucagon-like peptide-1 receptor agonist (GLP-1RA), a novel hypoglycemic agent for the treatment of type 2 diabetes, has well-known effects such as lowering blood sugar, ameliorating inflammation, reducing weight, and lowering blood lipids. It has also been shown that it can influence the proliferation and survival of cells and has a certain effect on the prognosis of some neoplastic diseases. In this study, the potential effects of GLP-1RAs on the occurrence and development of tumors were reviewed to provide new ideas for the prevention and treatment of tumors in patients.
Collapse
Affiliation(s)
- Lisan Ji
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Xianzhen He
- Children’s Medical Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xinwen Min
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Handong Yang
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Wenwen Wu
- School of Public Health, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hao Xu
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Jun Chen
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
- Virology Key Laboratory of Shiyan City, Hubei University of Medicine, Shiyan, China
| | - Aihua Mei
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
49
|
Chen Z, Deng X, Shi C, Jing H, Tian Y, Zhong J, Chen G, Xu Y, Luo Y, Zhu Y. GLP-1R-positive neurons in the lateral septum mediate the anorectic and weight-lowering effects of liraglutide in mice. J Clin Invest 2024; 134:e178239. [PMID: 39225090 PMCID: PMC11364389 DOI: 10.1172/jci178239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Liraglutide, a glucagon-like peptide-1 (GLP-1) analog, is approved for obesity treatment, but the specific neuronal sites that contribute to its therapeutic effects remain elusive. Here, we show that GLP-1 receptor-positive (GLP-1R-positive) neurons in the lateral septum (LSGLP-1R) play a critical role in mediating the anorectic and weight-loss effects of liraglutide. LSGLP-1R neurons were robustly activated by liraglutide, and chemogenetic activation of these neurons dramatically suppressed feeding. Targeted knockdown of GLP-1 receptors within the LS, but not in the hypothalamus, substantially attenuated liraglutide's ability to inhibit feeding and lower body weight. The activity of LSGLP-1R neurons rapidly decreased during naturalistic feeding episodes, while synaptic inactivation of LSGLP-1R neurons diminished the anorexic effects triggered by liraglutide. Together, these findings offer critical insights into the functional role of LSGLP-1R neurons in the physiological regulation of energy homeostasis and delineate their instrumental role in mediating the pharmacological efficacy of liraglutide.
Collapse
Affiliation(s)
- Zijun Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaofei Deng
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cuijie Shi
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Haiyang Jing
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yu Tian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Jiafeng Zhong
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Gaowei Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yunlong Xu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Yixiao Luo
- Hunan Province People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
50
|
Park S, Kim EK. Machine Learning-Based Plasma Metabolomics in Liraglutide-Treated Type 2 Diabetes Mellitus Patients and Diet-Induced Obese Mice. Metabolites 2024; 14:483. [PMID: 39330490 PMCID: PMC11434292 DOI: 10.3390/metabo14090483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Liraglutide, a glucagon-like peptide-1 receptor agonist, is effective in the treatment of type 2 diabetes mellitus (T2DM) and obesity. Despite its benefits, including improved glycemic control and weight loss, the common metabolic changes induced by liraglutide and correlations between those in rodents and humans remain unknown. Here, we used advanced machine learning techniques to analyze the plasma metabolomic data in diet-induced obese (DIO) mice and patients with T2DM treated with liraglutide. Among the machine learning models, Support Vector Machine was the most suitable for DIO mice, and Gradient Boosting was the most suitable for patients with T2DM. Through the cross-evaluation of machine learning models, we found that liraglutide promotes metabolic shifts and interspecies correlations in these shifts between DIO mice and patients with T2DM. Our comparative analysis helped identify metabolic correlations influenced by liraglutide between humans and rodents and may guide future therapeutic strategies for T2DM and obesity.
Collapse
Affiliation(s)
- Seokjae Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea;
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Eun-Kyoung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea;
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| |
Collapse
|