1
|
Ajoolabady A, Pratico D, Mazidi M, Davies IG, Lip GYH, Seidah N, Libby P, Kroemer G, Ren J. PCSK9 in metabolism and diseases. Metabolism 2025; 163:156064. [PMID: 39547595 DOI: 10.1016/j.metabol.2024.156064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/02/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
PCSK9 is a serine protease that regulates plasma levels of low-density lipoprotein (LDL) and cholesterol by mediating the endolysosomal degradation of LDL receptor (LDLR) in the liver. When PCSK9 functions unchecked, it leads to increased degradation of LDLR, resulting in elevated circulatory levels of LDL and cholesterol. This dysregulation contributes to lipid and cholesterol metabolism abnormalities, foam cell formation, and the development of various diseases, including cardiovascular disease (CVD), viral infections, cancer, and sepsis. Emerging clinical and experimental evidence highlights an imperative role for PCSK9 in metabolic anomalies such as hypercholesterolemia and hyperlipidemia, as well as inflammation, and disturbances in mitochondrial homeostasis. Moreover, metabolic hormones - including insulin, glucagon, adipokines, natriuretic peptides, and sex steroids - regulate the expression and circulatory levels of PCSK9, thus influencing cardiovascular and metabolic functions. In this comprehensive review, we aim to elucidate the regulatory role of PCSK9 in lipid and cholesterol metabolism, pathophysiology of diseases such as CVD, infections, cancer, and sepsis, as well as its pharmaceutical and non-pharmaceutical targeting for therapeutic management of these conditions.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Mohsen Mazidi
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK; King's College London, Department of Twin Research & Genetic Epidemiology, South Wing St Thomas', London, UK; Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ian G Davies
- School of Sport and Exercise Sciences, Faculty of Science, Liverpool John Moores University, Copperas Hill, Liverpool L3 5AJ, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Nabil Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM, affiliated to the University of Montreal), Montreal, QC H2W 1R7, Canada.
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
2
|
Moreno-Lopez M, Louvet I, Delalleau N, Acosta-Montalvo A, Thevenet J, Pasquetti G, Gmyr V, Kerr-Conte J, Pattou F, Bonner C, Saponaro C. The role of the glucagon-FGF21 axis in improving beta cell function during glucose intolerance and SGLT2 inhibition. Diabetes Obes Metab 2025; 27:885-898. [PMID: 39618173 DOI: 10.1111/dom.16089] [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: 07/25/2024] [Revised: 10/30/2024] [Accepted: 11/09/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVE Although primarily secreted by the liver, Fibroblast Growth Factor 21 (FGF21) is also expressed in the pancreas, where its function remains unclear. This study aims to elucidate the role of the glucagon-FGF21 interaction in the metabolic benefits of SGLT2 inhibition (SGLT2i) and hypothesizes it is key to enhancing glucose and lipid metabolism in individuals with glucose intolerance or type 2 diabetes (T2D). METHODS FGF21, FGF1R, and β-klotho expression in human pancreas was analysed by RNAscope, qPCR and immunofluorescent techniques. Glucose-stimulated insulin secretion (GSIS) assay was used to investigate the effects of recombinant FGF21 (rFGF21) on islets from donors with glucose intolerance or T2D. To explore the role of the glucagon-FGF21 axis in the benefits of SGLT2i, we used WT and Sglt2 knockout (KO) mice fed a chow diet (CD) or a high-fat diet (HFD) and chronically treated with vehicle or dapagliflozin. RESULTS Chronic rFGF21 treatment enhanced GSIS in islets from donors with glucose intolerance, with increased FGFR1 expression, suggesting FGF21's greater efficacy in the early stages of disease. In diet-induced insulin-resistant mice, dapagliflozin reduced postprandial glycaemia and elevated plasma glucagon and FGF21 levels. Sglt2 KO mice on a CD showed increased fasting plasma glucagon without changes in FGF21. In diet-induced insulin-resistant Sglt2 KO mice, elevated glucagon and FGF21 levels paralleled chronic dapagliflozin treatment, indicating similar metabolic adaptations in both models. CONCLUSION Our findings indicate FGF21 as a crucial mediator in liver-pancreas crosstalk, improving lipid and glucose metabolism, enhancing pancreatic function, and potentiating the therapeutic efficacy of SGLT2i, thereby representing a target for prediabetes treatment.
Collapse
Affiliation(s)
- Maria Moreno-Lopez
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Isaline Louvet
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Nathalie Delalleau
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Ana Acosta-Montalvo
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Julien Thevenet
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Gianni Pasquetti
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Valery Gmyr
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Julie Kerr-Conte
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Francois Pattou
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Caroline Bonner
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Chiara Saponaro
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
3
|
Mehdi SF, Qureshi MH, Pervaiz S, Kumari K, Saji E, Shah M, Abdullah A, Zahoor K, Qadeer HA, Katari DK, Metz C, Mishra L, LeRoith D, Tracey K, Brownstein MJ, Roth J. Endocrine and metabolic alterations in response to systemic inflammation and sepsis: a review article. Mol Med 2025; 31:16. [PMID: 39838305 PMCID: PMC11752782 DOI: 10.1186/s10020-025-01074-z] [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: 10/17/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025] Open
Abstract
Severe sepsis is cognate with life threatening multi-organ dysfunction. There is a disturbance in endocrine functions with alterations in several hormonal pathways. It has frequently been linked with dysfunction in the hypothalamic pituitary-adrenal axis (HPA). Increased cortisol or cortisolemia is evident throughout the acute phase, along with changes in the hypothalamic pituitary thyroid (HPT) axis, growth hormone-IGF-1 axis, insulin-glucose axis, leptin, catecholamines, renin angiotensin aldosterone axis, ghrelin, glucagon, hypothalamic pituitary gonadal (HGA) axis, and fibroblast growth factor-21. These changes and metabolic alterations constitute the overall response to infection in sepsis. Further research is essential to look into the hormonal changes that occur during sepsis, not only to understand their potential relevance in therapy but also because they may serve as prognostic indicators.
Collapse
Affiliation(s)
- Syed Faizan Mehdi
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | | | - Salman Pervaiz
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Karishma Kumari
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Edwin Saji
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Mahnoor Shah
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Ahmad Abdullah
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Kamran Zahoor
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Hafiza Amna Qadeer
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Disha Kumari Katari
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Christine Metz
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Lopa Mishra
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes & Bone Disease, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Kevin Tracey
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA
| | | | - Jesse Roth
- The Feinstein Institutes for Medical Research/Northwell Health, Manhasset, NY, USA.
| |
Collapse
|
4
|
Šestan M, Raposo B, Rendas M, Brea D, Pirzgalska R, Rasteiro A, Aliseychik M, Godinho I, Ribeiro H, Carvalho T, Wueest S, Konrad D, Veiga-Fernandes H. Neuronal-ILC2 interactions regulate pancreatic glucagon and glucose homeostasis. Science 2025; 387:eadi3624. [PMID: 39818880 DOI: 10.1126/science.adi3624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/02/2024] [Accepted: 11/08/2024] [Indexed: 01/19/2025]
Abstract
The immune system shapes body metabolism, while interactions between peripheral neurons and immune cells control tissue homeostasis and immunity. However, whether peripheral neuroimmune interactions orchestrate endocrine system functions remains unexplored. After fasting, mice lacking type 2 innate lymphoid cells (ILC2s) displayed disrupted glucose homeostasis, impaired pancreatic glucagon secretion, and inefficient hepatic gluconeogenesis. Additionally, intestinal ILC2s were found in the pancreas, which was dependent on their expression of the adrenergic beta 2 receptor. Targeted activation of catecholaminergic intestinal neurons promoted the accumulation of ILC2s in the pancreas. Our work provides evidence that immune cells can be regulated by neuronal signals in response to fasting, activating an inter-organ communication route that promotes pancreatic endocrine function and regulation of blood glucose levels.
Collapse
Affiliation(s)
- Marko Šestan
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Bruno Raposo
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Miguel Rendas
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - David Brea
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Roksana Pirzgalska
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Ana Rasteiro
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Maria Aliseychik
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Inês Godinho
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Hélder Ribeiro
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Tania Carvalho
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, University of Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, University of Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Henrique Veiga-Fernandes
- Champalimaud Foundation. Champalimaud Centre for the Unknown. Champalimaud Research. Lisbon, Portugal
| |
Collapse
|
5
|
Wharton S, le Roux CW, Kosiborod MN, Platz E, Brueckmann M, Jastreboff AM, Ajaz Hussain S, Pedersen SD, Borowska L, Unseld A, Kloer IM, Kaplan LM. Survodutide for treatment of obesity: rationale and design of two randomized phase 3 clinical trials (SYNCHRONIZE™-1 and -2). Obesity (Silver Spring) 2025; 33:67-77. [PMID: 39495965 DOI: 10.1002/oby.24184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 11/06/2024]
Abstract
OBJECTIVE The objective of this study was to describe the rationale and design of two multinational phase 3 clinical trials of survodutide, an investigational glucagon and glucagon-like peptide-1 receptor dual agonist for the treatment of obesity with or without type 2 diabetes (T2D; SYNCHRONIZE-1 and -2). METHODS In these ongoing double-blind trials, participants were randomized to once-weekly subcutaneous injections of survodutide or placebo added to lifestyle modification. Survodutide doses are uptitrated to 3.6 or 6.0 mg, and dose flexibility is permitted. Participants (n = 726) in SYNCHRONIZE-1 (NCT06066515) have a baseline BMI ≥ 30 kg/m2 or ≥27 kg/m2 with at least one obesity-related complication but without T2D; participants (n = 755) in SYNCHRONIZE-2 (NCT06066528) have a baseline BMI ≥ 27 kg/m2 and T2D. The primary endpoints are percentage change in body weight and proportion of participants achieving ≥5% body weight reduction from baseline to week 76. Secondary endpoints include change in systolic blood pressure and measures of glycemia. A SYNCHRONIZE-1 substudy is evaluating changes in body composition and liver fat content using magnetic resonance imaging. CONCLUSIONS These trials are designed to provide robust evaluation of the efficacy, safety, and tolerability of survodutide for the treatment of obesity in the presence or absence of T2D.
Collapse
Affiliation(s)
- Sean Wharton
- McMaster University, Hamilton, Ontario and University of Toronto, Toronto, Ontario, Canada
| | - Carel W le Roux
- St. Vincent's University Hospital and University College Dublin School of Medicine, Dublin, Ireland
| | - Mikhail N Kosiborod
- Saint Luke's Mid America Heart Institute and University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Elke Platz
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Martina Brueckmann
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ania M Jastreboff
- Section of Endocrinology (Internal Medicine & Pediatrics) and Yale Obesity Research Center (Y-Weight), Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Sue D Pedersen
- C-ENDO Diabetes and Endocrinology Clinic and University of Calgary, Calgary, Alberta, Canada
| | - Luiza Borowska
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Anna Unseld
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | - Isabel M Kloer
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Lee M Kaplan
- Section of Obesity Medicine and Center for Digestive Health, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
6
|
Long A, Wang Y, Guo Y, Hong J, Ning G, Meng Z, Wang J, Wang Y. A famsin-glucagon axis mediates glucose homeostasis. Cell Metab 2024:S1550-4131(24)00454-6. [PMID: 39706194 DOI: 10.1016/j.cmet.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/31/2024] [Accepted: 11/12/2024] [Indexed: 12/23/2024]
Abstract
Glucagon is essential for glucose homeostasis, and its dysregulation is associated with diabetes. Despite extensive research, the mechanisms governing glucagon secretion remain incompletely understood. Here, we unveil that famsin, a gut-secreted hormone, promotes glucagon release and modulates glucose homeostasis. Mechanistically, famsin binds to its receptor OLFR796 in mice (OR10P1 in humans), initiating calcium release in the endoplasmic reticulum of islet α cells. This process triggers glucagon secretion, consequently promoting hepatic glucose production through glucagon signaling. Furthermore, deficiency of famsin signaling reduces hepatic glucose production and lowers blood glucose levels, underscoring the significance of the famsin-glucagon axis in glucose homeostasis. Therefore, our findings establish famsin as a crucial regulator of glucagon secretion and provide valuable insights into the intricate gut-islet-liver interorgan crosstalk that maintains glucose homeostasis.
Collapse
Affiliation(s)
- Aijun Long
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China; Metabolic Syndrome Research Center, Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yazhuo Wang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yihua Guo
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Zhuoxian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China.
| | - Yiguo Wang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
7
|
Kaya E, Yilmaz Y, Alkhouri N. Survodutide in MASH: bridging the gap between hepatic and systemic metabolic dysfunction. Expert Opin Investig Drugs 2024; 33:1167-1176. [PMID: 39663847 DOI: 10.1080/13543784.2024.2441865] [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: 07/21/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION Glucagon-like peptide-1 receptor (GLP-1 R) agonists have demonstrated remarkable effectiveness in the treatment of obesity and type 2 diabetes. Although these agents provide beneficial effects for metabolic dysfunction-associated steatohepatitis (MASH) through their glucose-lowering and weight-reducing properties, their efficacy in promoting fibrosis regression remains unproven. Survodutide, an investigational dual agonist that simultaneously targets both the glucagon receptor (GCGR) and GLP-1 R, has emerged as a promising therapeutic candidate for the comprehensive management of obesity and MASH. By engaging these two critical receptors, this drug has the potential to offer a broad spectrum of metabolic benefits, addressing multiple pathogenic mechanisms underlying these interrelated disorders. AREAS COVERED This review examines the pharmacological profile, clinical efficacy, and safety data of survodutide derived from phase 1 and 2 clinical trials. EXPERT OPINION Survodutide's dual agonism of the GCGR and GLP-1 R may surpass the efficacy of selective GLP-1 R agonists, demonstrating significant potential in resolving MASH and promoting fibrosis regression. The drug is generally well tolerated, with primarily manageable gastrointestinal adverse effects. As survodutide progresses through phase 3 clinical development, its potential to provide a more effective and holistic approach to treating MASH and its comorbidities may significantly improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Eda Kaya
- Department of Medicine, Knappschaftskrankenhaus Bochum, Ruhr University, Bochum, Germany
- Department of Hepatology, The Global NASH Council, Washington, DC, USA
| | - Yusuf Yilmaz
- Department of Hepatology, The Global NASH Council, Washington, DC, USA
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdogan University, Rize, Türkiye
| | - Naim Alkhouri
- Department of Hepatology, The Global NASH Council, Washington, DC, USA
- Department of Hepatology, Arizona Liver Health, Chandler, Arizona, USA
| |
Collapse
|
8
|
Schuppelius B, Schüler R, Pivovarova-Ramich O, Hornemann S, Busjahn A, Machann J, Kruse M, Park SQ, Kabisch S, Csanalosi M, Ost AC, Pfeiffer AFH. Alterations in Glucagon Levels and the Glucagon-to-Insulin Ratio in Response to High Dietary Fat or Protein Intake in Healthy Lean Adult Twins: A Post Hoc Analysis. Nutrients 2024; 16:3905. [PMID: 39599691 PMCID: PMC11597242 DOI: 10.3390/nu16223905] [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: 10/14/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Emerging data support evidence of the essential role of glucagon for lipid metabolism. However, data on the role of dietary fat intake for glucagon secretion is limited. This analysis investigated whether altering nutritional fat intake affects glucagon levels in healthy subjects. Methods: A total of 92 twins (age: 31 ± 14 years, BMI: 23 ± 3 kg/m2) consumed two 6-week diets: first a low-fat, high-carbohydrate diet (LFD) followed by an isocaloric high-fat, low-carbohydrate diet (HFD). In total, 24 twins (age: 39 ± 15 years, BMI: 24 ± 2 kg/m2) continued with a high-protein diet (HPD). Clinical investigations were performed after 6 weeks of the LFD, after 1 and 6 weeks of the HFD and after 6 weeks of the HPD. Results: The LFD caused a significant decrease in fasting glucagon (-27%, p < 0.001) compared to baseline. After 6 weeks of the HFD, glucagon increased (117%, p < 0.001 vs. LFD), while free fatty acids decreased. Six weeks of the HPD further increased glucagon levels (72%, p = 0.502 vs. HFD), although fasting amino acid levels remained constant. Fasting insulin and HOMA-IR moderately increased after one week of the HFD, while six weeks of the HPD significantly decreased both. The fasting glucagon-to-insulin ratio decreased during the LFD (p < 0.001) but increased after the HFD (p < 0.001) and even further increased after the HPD (p = 0.018). Liver fat, triglycerides and blood glucose did not increase during the HFD. The heritability of glucagon levels was 45% with the LFD. Conclusions: An HFD increases glucagon levels and the glucagon-to-insulin ratio under isocaloric conditions compared to an LFD in healthy lean subjects. This rise in glucagon may represent a metabolic response to prevent hepatic steatosis, as glucagon increases have been previously shown to induce hepatic fat oxidation.
Collapse
Affiliation(s)
- Bettina Schuppelius
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Rita Schüler
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Olga Pivovarova-Ramich
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Department of Molecular Metabolism and Precision Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| | - Silke Hornemann
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Andreas Busjahn
- HealthTwiSt GmbH, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, University of Tübingen, Geissweg 3, 72076 Tübingen, Germany
| | - Michael Kruse
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Soyoung Q. Park
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
- Department of Decision Neuroscience and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Neuroscience Research Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Stefan Kabisch
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| | - Marta Csanalosi
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Anne-Cathrin Ost
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| |
Collapse
|
9
|
Lawitz EJ, Fraessdorf M, Neff GW, Schattenberg JM, Noureddin M, Alkhouri N, Schmid B, Andrews CP, Takács I, Hussain SA, Fenske WK, Gane EJ, Hosseini-Tabatabaei A, Sanyal AJ, Mazo DF, Younes R. Efficacy, tolerability and pharmacokinetics of survodutide, a glucagon/glucagon-like peptide-1 receptor dual agonist, in cirrhosis. J Hepatol 2024; 81:837-846. [PMID: 38857788 DOI: 10.1016/j.jhep.2024.06.003] [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: 04/25/2024] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND & AIMS Survodutide is a glucagon/glucagon-like peptide-1 receptor dual agonist in development for the treatment of metabolic dysfunction-associated steatohepatitis (MASH). We investigated the pharmacokinetic and safety profile of survodutide in people with cirrhosis. METHODS This multinational, non-randomized, open-label, phase I clinical trial initially evaluated a single subcutaneous dose of survodutide 0.3 mg in people with Child-Pugh class A, B or C cirrhosis and healthy individuals with or without overweight/obesity matched for age, sex, and weight; the primary endpoints were the area under the plasma concentration-time curve from 0 to infinity (AUC0-∞) and maximal plasma concentration (Cmax). Subsequently, people with overweight/obesity with or without cirrhosis (Child-Pugh class A or B) received once-weekly subcutaneous doses escalated from 0.3 mg to 6.0 mg over 24 weeks then maintained for 4 weeks; the primary endpoint was drug-related treatment-emergent adverse events, with MASH/cirrhosis-related endpoints explored. RESULTS In the single-dose cohorts (n = 41), mean AUC0-∞ and Cmax were similar in those with cirrhosis compared with healthy individuals (90% CIs for adjusted geometric mean ratios spanned 1). Drug-related adverse events occurred in 25.0% of healthy individuals and ≤25.0% of those with cirrhosis after single doses, and 82.4% and 87.5%, respectively, of the multiple-dose cohorts (n = 41) over 28 weeks. Liver fat content, liver stiffness, liver volume, body weight, and other hepatic and metabolic disease markers were generally reduced after 28 weeks of survodutide treatment. CONCLUSIONS Survodutide is generally tolerable in people with compensated or decompensated cirrhosis, does not require pharmacokinetic-related dose adjustment, and may improve liver-related non-invasive tests, supporting its investigation for MASH-related cirrhosis. IMPACT AND IMPLICATIONS Survodutide is a glucagon receptor/glucagon-like peptide-1 receptor dual agonist in development for treatment of metabolic dysfunction-associated steatohepatitis (MASH), which causes cirrhosis in ∼20% of cases. This trial delineates the pharmacokinetic and safety profile of survodutide in people with compensated or decompensated cirrhosis, and revealed associated reductions in liver fat content, markers of liver fibrosis and body weight. These findings have potential relevance for people with MASH-including those with decompensated cirrhosis, who are usually excluded from clinical trials of investigational drugs. Based on this study, further investigation of survodutide for MASH-related cirrhosis is warranted. CLINICALTRIALS GOV IDENTIFIER NCT05296733.
Collapse
Affiliation(s)
- Eric J Lawitz
- Texas Liver Institute, University of Texas Health San Antonio, San Antonio, TX, USA.
| | | | - Guy W Neff
- Covenant Metabolic Specialists, Sarasota and Fort Myers, FL, USA
| | - Jörn M Schattenberg
- Department of Internal Medicine II, University Medical Center Homburg, Homburg and Saarland University, Saarbrücken, Germany
| | - Mazen Noureddin
- Houston Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Naim Alkhouri
- Hepatology Division, Arizona Liver Health, Phoenix, AZ, USA
| | | | | | - István Takács
- Department of Internal Medicine and Oncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | | | - Wiebke K Fenske
- Department of Internal Medicine I, Endocrinology, Diabetes and Metabolism, Bergmannsheil University Hospital Bochum, Bochum, Germany
| | - Edward J Gane
- New Zealand Liver Transplant Unit, Auckland City Hospital and University of Auckland, Auckland, New Zealand
| | | | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | | | | |
Collapse
|
10
|
Holst JJ. GLP-1 physiology in obesity and development of incretin-based drugs for chronic weight management. Nat Metab 2024; 6:1866-1885. [PMID: 39160334 DOI: 10.1038/s42255-024-01113-9] [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: 01/30/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024]
Abstract
The introduction of the highly potent incretin receptor agonists semaglutide and tirzepatide has marked a new era in the treatment of type 2 diabetes and obesity. With normalisation of glycated haemoglobin levels and weight losses around 15-25%, therapeutic goals that were previously unrealistic are now within reach, and clinical trials have documented that these effects are associated with reduced risk of cardiovascular events and premature mortality. Here, I review this remarkable development from the earliest observations of glucose lowering and modest weight losses with native glucagon-like peptide (GLP)-1 and short acting compounds, to the recent development of highly active formulations and new molecules. I will classify these agents as GLP-1-based therapies in the understanding that these compounds or combinations may have actions on other receptors as well. The physiology of GLP-1 is discussed as well as its mechanisms of actions in obesity, in particular, the role of sensory afferents and GLP-1 receptors in the brain. I provide details regarding the development of GLP-1 receptor agonists for anti-obesity therapy and discuss the possible mechanism behind their beneficial effects on adverse cardiovascular events. Finally, I highlight new pharmacological developments, including oral agents, and discuss important questions regarding maintenance therapy.
Collapse
Affiliation(s)
- Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences. Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
11
|
Ansari S, Khoo B, Tan T. Targeting the incretin system in obesity and type 2 diabetes mellitus. Nat Rev Endocrinol 2024; 20:447-459. [PMID: 38632474 DOI: 10.1038/s41574-024-00979-9] [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] [Accepted: 03/12/2024] [Indexed: 04/19/2024]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are widespread, non-communicable diseases that are responsible for considerable levels of morbidity and mortality globally, primarily in the form of cardiovascular disease (CVD). Changes to lifestyle and behaviour have insufficient long-term efficacy in most patients with these diseases; metabolic surgery, although effective, is not practically deliverable on the scale that is required. Over the past two decades, therapies based on incretin hormones, spearheaded by glucagon-like peptide 1 (GLP1) receptor agonists (GLP1RAs), have become the treatment of choice for obesity and T2DM, and clinical evidence now suggests that these agents have benefits for CVD. We review the latest advances in incretin-based pharmacotherapy. These include 'GLP1 plus' agents, which combine the known advantages of GLP1RAs with the activity of additional hormones, such as glucose-dependent insulinotropic peptide, glucagon and amylin, to achieve desired therapeutic goals. Second-generation non-peptidic oral GLP1RAs promise to extend the benefits of GLP1 therapy to those who do not want, or cannot have, subcutaneous injection therapy. We conclude with a discussion of the knowledge gaps that must be addressed before incretin-based therapies can be properly deployed for maximum benefit in the treatment of obesity and T2DM.
Collapse
Affiliation(s)
- Saleem Ansari
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Bernard Khoo
- Department of Endocrinology, Division of Medicine, Royal Free Campus, University College London, London, UK
| | - Tricia Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK.
| |
Collapse
|
12
|
Kusminski CM, Perez-Tilve D, Müller TD, DiMarchi RD, Tschöp MH, Scherer PE. Transforming obesity: The advancement of multi-receptor drugs. Cell 2024; 187:3829-3853. [PMID: 39059360 PMCID: PMC11286204 DOI: 10.1016/j.cell.2024.06.003] [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: 11/09/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024]
Abstract
For more than a century, physicians have searched for ways to pharmacologically reduce excess body fat. The tide has finally turned with recent advances in biochemically engineered agonists for the receptor of glucagon-like peptide-1 (GLP-1) and their use in GLP-1-based polyagonists. These polyagonists reduce body weight through complementary pharmacology by incorporating the receptors for glucagon and/or the glucose-dependent insulinotropic polypeptide (GIP). In their most advanced forms, gut-hormone polyagonists achieve an unprecedented weight reduction of up to ∼20%-30%, offering a pharmacological alternative to bariatric surgery. Along with favorable effects on glycemia, fatty liver, and kidney disease, they also offer beneficial effects on the cardiovascular system and adipose tissue. These new interventions, therefore, hold great promise for the future of anti-obesity medications.
Collapse
Affiliation(s)
- Christine M Kusminski
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Munich, Germany; German Center for Diabetes Research (DZD) and Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | | | - Matthias H Tschöp
- Helmholtz Munich, Munich, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
13
|
McGlone ER, Hope DCD, Davies I, Dore M, Goldin R, Jones B, Liu Z, Li JV, Vorkas PA, Khoo B, Carling D, Minnion J, Bloom SR, Tan TMM. Chronic treatment with glucagon-like peptide-1 and glucagon receptor co-agonist causes weight loss-independent improvements in hepatic steatosis in mice with diet-induced obesity. Biomed Pharmacother 2024; 176:116888. [PMID: 38861859 DOI: 10.1016/j.biopha.2024.116888] [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: 05/03/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
OBJECTIVES Co-agonists at the glucagon-like peptide-1 and glucagon receptors (GLP1R/GCGR) show promise as treatments for metabolic dysfunction-associated steatotic liver disease (MASLD). Although most co-agonists to date have been heavily GLP1R-biased, glucagon directly acts on the liver to reduce fat content. The aims of this study were to investigate a GCGR-biased co-agonist as treatment for hepatic steatosis in mice. METHODS Mice with diet-induced obesity (DIO) were treated with Dicretin, a GLP1/GCGR co-agonist with high potency at the GCGR, Semaglutide (GLP1R monoagonist) or food restriction over 24 days, such that their weight loss was matched. Hepatic steatosis, glucose tolerance, hepatic transcriptomics, metabolomics and lipidomics at the end of the study were compared with Vehicle-treated mice. RESULTS Dicretin lead to superior reduction of hepatic lipid content when compared to Semaglutide or equivalent weight loss by calorie restriction. Markers of glucose tolerance and insulin resistance improved in all treatment groups. Hepatic transcriptomic and metabolomic profiling demonstrated many changes that were unique to Dicretin-treated mice. These include some known targets of glucagon signaling and others with as yet unclear physiological significance. CONCLUSIONS Our study supports the development of GCGR-biased GLP1/GCGR co-agonists for treatment of MASLD and related conditions.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Department of Surgery and Cancer, Imperial College London, London, UK; Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - David C D Hope
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Iona Davies
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Marian Dore
- Genomics facility, MRC Laboratory of Medical Sciences (LMS), Imperial College London, London, UK
| | - Rob Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ben Jones
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Zhigang Liu
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jia V Li
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Panagiotis A Vorkas
- Institute of Applied Biosciences, Centre for Research and Technology Hellas (INAB|CERTH), Thessaloniki 57001, Greece; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | - Bernard Khoo
- Endocrinology, Division of Medicine, University College London, London, UK
| | - David Carling
- Cellular Stress group, MRC LMS, Imperial College London, London, UK
| | - James Minnion
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stephen R Bloom
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M-M Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
14
|
Kajani S, Laker RC, Ratkova E, Will S, Rhodes CJ. Hepatic glucagon action: beyond glucose mobilization. Physiol Rev 2024; 104:1021-1060. [PMID: 38300523 DOI: 10.1152/physrev.00028.2023] [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: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Glucagon's ability to promote hepatic glucose production has been known for over a century, with initial observations touting this hormone as a diabetogenic agent. However, glucagon receptor agonism [when balanced with an incretin, including glucagon-like peptide 1 (GLP-1) to dampen glucose excursions] is now being developed as a promising therapeutic target in the treatment of metabolic diseases, like metabolic dysfunction-associated steatotic disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH), and may also have benefit for obesity and chronic kidney disease. Conventionally regarded as the opposing tag-team partner of the anabolic mediator insulin, glucagon is gradually emerging as more than just a "catabolic hormone." Glucagon action on glucose homeostasis within the liver has been well characterized. However, growing evidence, in part thanks to new and sensitive "omics" technologies, has implicated glucagon as more than just a "glucose liberator." Elucidation of glucagon's capacity to increase fatty acid oxidation while attenuating endogenous lipid synthesis speaks to the dichotomous nature of the hormone. Furthermore, glucagon action is not limited to just glucose homeostasis and lipid metabolism, as traditionally reported. Glucagon plays key regulatory roles in hepatic amino acid and ketone body metabolism, as well as mitochondrial turnover and function, indicating broader glucagon signaling consequences for metabolic homeostasis mediated by the liver. Here we examine the broadening role of glucagon signaling within the hepatocyte and question the current dogma, to appreciate glucagon as more than just that "catabolic hormone."
Collapse
Affiliation(s)
- Sarina Kajani
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Rhianna C Laker
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Ekaterina Ratkova
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sarah Will
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Christopher J Rhodes
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| |
Collapse
|
15
|
Karimi R, Yanovich A, Elbarbry F, Cleven A. Adaptive Effects of Endocrine Hormones on Metabolism of Macronutrients during Fasting and Starvation: A Scoping Review. Metabolites 2024; 14:336. [PMID: 38921471 PMCID: PMC11205672 DOI: 10.3390/metabo14060336] [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: 03/29/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Food deprivation can occur for different reasons. Fasting (<24 h duration) occurs to meet religious or well-being goals. Starvation (>1-day duration) occurs when there is intentional (hunger strike or treatment of a medical condition) or unintentional (anorexia nervosa, drought, epidemic famine, war, or natural disaster) food deprivation. A scoping review was undertaken using the PubMed database to explore 1805 abstracts and review 88 eligible full-text articles to explore the adaptive relationships that emerge between cortisol, insulin, glucagon, and thyroid hormones on the metabolic pathways of macronutrients in humans during fasting and starvation. The collected data indicate that fasting and starvation prime the human body to increase cortisol levels and decrease the insulin/glucagon ratio and triiodothyronine (T3) levels. During fasting, increased levels of cortisol and a decreased insulin/glucagon ratio enhance glycogenolysis and reduce the peripheral uptake of glucose and glycogenesis, whereas decreased T3 levels potentially reduce glycogenolysis. During starvation, increased levels of cortisol and a decreased insulin/glucagon ratio enhance lipolysis, proteolysis, fatty acid and amino acid oxidation, ketogenesis, and ureagenesis, and decreased T3 levels reduce thermogenesis. We present a potential crosstalk between T3 and the above hormones, including between T3 and leptin, to extend their adaptive roles in the metabolism of endogenous macronutrients during food deprivation.
Collapse
Affiliation(s)
- Reza Karimi
- Pacific University School of Pharmacy, 222 SE 8th Avenue, HPC-Ste 451, Hillsboro, OR 97123, USA; (A.Y.); (F.E.); (A.C.)
| | | | | | | |
Collapse
|
16
|
Rix I, Lund AB, Garvey LF, Hansen CP, Chabanova E, Hartmann B, Holst JJ, Vilsbøll T, Van Hall G, Knop FK. Increased hepatic glucagon sensitivity in totally pancreatectomised patients. Eur J Endocrinol 2024; 190:446-457. [PMID: 38781444 DOI: 10.1093/ejendo/lvae054] [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: 11/16/2023] [Revised: 04/02/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE The metabolic phenotype of totally pancreatectomised patients includes hyperaminoacidaemia and predisposition to hypoglycaemia and hepatic lipid accumulation. We aimed to investigate whether the loss of pancreatic glucagon may be responsible for these changes. METHODS Nine middle-aged, normal-weight totally pancreatectomised patients, nine patients with type 1 diabetes (C-peptide negative), and nine matched controls underwent two separate experimental days, each involving a 150-min intravenous infusion of glucagon (4 ng/kg/min) or placebo (saline) under fasting conditions while any basal insulin treatment was continued. RESULTS Glucagon infusion increased plasma glucagon to similar high physiological levels in all groups. The infusion increased hepatic glucose production and decreased plasma concentration of most amino acids in all groups, with more pronounced effects in the totally pancreatectomised patients compared with the other groups. Glucagon infusion diminished fatty acid re-esterification and tended to decrease plasma concentrations of fatty acids in the totally pancreatectomised patients but not in the type 1 diabetes patients. CONCLUSION Totally pancreatectomised patients were characterised by increased sensitivity to exogenous glucagon at the level of hepatic glucose, amino acid, and lipid metabolism, suggesting that the metabolic disturbances characterising these patients may be rooted in perturbed hepatic processes normally controlled by pancreatic glucagon.
Collapse
Affiliation(s)
- Iben Rix
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, 2900 Hellerup, Denmark
- Medical & Science, Zealand Pharma A/S, 2860 Søborg, Denmark
| | - Asger B Lund
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, 2900 Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lars F Garvey
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, 2900 Hellerup, Denmark
| | - Carsten P Hansen
- Department of Surgery, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| | - Elizaveta Chabanova
- Department of Radiology, Copenhagen University Hospital - Herlev and Gentofte, 2730 Herlev, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, 2900 Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Gerrit Van Hall
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Clinical Metabolomics Core Facility, Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, 2100 Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, 2900 Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
17
|
McGlone ER, Tan TMM. Glucagon-based therapy for people with diabetes and obesity: What is the sweet spot? Peptides 2024; 176:171219. [PMID: 38615717 DOI: 10.1016/j.peptides.2024.171219] [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: 03/06/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
People with obesity and type 2 diabetes have a high prevalence of metabolic-associated steatotic liver disease, hyperlipidemia and cardiovascular disease. Glucagon increases hepatic glucose production; it also decreases hepatic fat accumulation, improves lipidemia and increases energy expenditure. Pharmaceutical strategies to antagonize the glucagon receptor improve glycemic outcomes in people with diabetes and obesity, but they increase hepatic steatosis and worsen dyslipidemia. Co-agonism of the glucagon and glucagon-like peptide-1 (GLP-1) receptors has emerged as a promising strategy to improve glycemia in people with diabetes and obesity. Addition of glucagon receptor agonism enhances weight loss, reduces liver fat and ameliorates dyslipidemia. Prior to clinical use, however, further studies are needed to investigate the safety and efficacy of glucagon and GLP-1 receptor co-agonists in people with diabetes and obesity and related conditions, with specific concerns regarding a higher prevalence of gastrointestinal side effects, loss of muscle mass and increases in heart rate. Furthermore, co-agonists with differing ratios of glucagon:GLP-1 receptor activity vary in their clinical effect; the optimum balance is yet to be identified.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Department of Surgery and Cancer, Imperial College London, London, UK; Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M-M Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
18
|
McFarlin BE, Duffin KL, Konkar A. Incretin and glucagon receptor polypharmacology in chronic kidney disease. Am J Physiol Endocrinol Metab 2024; 326:E747-E766. [PMID: 38477666 PMCID: PMC11551006 DOI: 10.1152/ajpendo.00374.2023] [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: 11/13/2023] [Accepted: 03/10/2024] [Indexed: 03/14/2024]
Abstract
Chronic kidney disease is a debilitating condition associated with significant morbidity and mortality. In recent years, the kidney effects of incretin-based therapies, particularly glucagon-like peptide-1 receptor agonists (GLP-1RAs), have garnered substantial interest in the management of type 2 diabetes and obesity. This review delves into the intricate interactions between the kidney, GLP-1RAs, and glucagon, shedding light on their mechanisms of action and potential kidney benefits. Both GLP-1 and glucagon, known for their opposing roles in regulating glucose homeostasis, improve systemic risk factors affecting the kidney, including adiposity, inflammation, oxidative stress, and endothelial function. Additionally, these hormones and their pharmaceutical mimetics may have a direct impact on the kidney. Clinical studies have provided evidence that incretins, including those incorporating glucagon receptor agonism, are likely to exhibit improved kidney outcomes. Although further research is necessary, receptor polypharmacology holds promise for preserving kidney function through eliciting vasodilatory effects, influencing volume and electrolyte handling, and improving systemic risk factors.
Collapse
Affiliation(s)
- Brandon E McFarlin
- Lilly Research Laboratories, Lilly Corporate CenterIndianapolisIndianaUnited States
| | - Kevin L Duffin
- Lilly Research Laboratories, Lilly Corporate CenterIndianapolisIndianaUnited States
| | - Anish Konkar
- Lilly Research Laboratories, Lilly Corporate CenterIndianapolisIndianaUnited States
| |
Collapse
|
19
|
Heckmann ND, Palmer R, Mayfield CK, Gucev G, Lieberman JR, Hong K. Glucagon-Like Peptide Receptor-1 Agonists Used for Medically-Supervised Weight Loss in Patients With Hip and Knee Osteoarthritis: Critical Considerations for the Arthroplasty Surgeon. Arthroplast Today 2024; 27:101327. [PMID: 39071832 PMCID: PMC11282421 DOI: 10.1016/j.artd.2024.101327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/08/2023] [Accepted: 01/27/2024] [Indexed: 07/30/2024] Open
Abstract
Patients with morbid obesity and concomitant hip or knee osteoarthritis represent a challenging patient demographic to treat as these patients often present earlier in life, have more severe symptoms, and have worse surgical outcomes following total hip and total knee arthroplasty. Previously, bariatric and metabolic surgeries represented one of the few weight loss interventions that morbidly obese patients could undergo prior to total joint arthroplasty. However, data regarding the reduction in complications with preoperative bariatric surgery remain mixed. Glucagon-like peptide receptor-1 (GLP-1) agonists have emerged as an effective treatment option for obesity in patients with and without diabetes mellitus. Furthermore, recent data suggest these medications may serve as potential anti-inflammatory and disease-modifying agents for numerous chronic conditions, including osteoarthritis. This review will discuss the GLP-1 agonists and GLP-1/glucose-dependent insulinotropic polypeptide dual agonists currently available, along with GLP-1/glucose-dependent insulinotropic polypeptide/glucagon triple agonists presently being developed to address the obesity epidemic. Furthermore, this review will address the potential problem of GLP-1-related delayed gastric emptying and its impact on the timing of elective total joint arthroplasty. The review aims to provide arthroplasty surgeons with a primer for implementing this class of medication in their current and future practice, including perioperative instructions and perioperative safety considerations when treating patients taking these medications.
Collapse
Affiliation(s)
- Nathanael D. Heckmann
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Ryan Palmer
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Cory K. Mayfield
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Gligor Gucev
- Department of Anesthesiology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Jay R. Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Kurt Hong
- Center for Clinical Nutrition, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
20
|
Duan J, He XH, Li SJ, Xu HE. Cryo-electron microscopy for GPCR research and drug discovery in endocrinology and metabolism. Nat Rev Endocrinol 2024; 20:349-365. [PMID: 38424377 DOI: 10.1038/s41574-024-00957-1] [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] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors, with many GPCRs having crucial roles in endocrinology and metabolism. Cryogenic electron microscopy (cryo-EM) has revolutionized the field of structural biology, particularly regarding GPCRs, over the past decade. Since the first pair of GPCR structures resolved by cryo-EM were published in 2017, the number of GPCR structures resolved by cryo-EM has surpassed the number resolved by X-ray crystallography by 30%, reaching >650, and the number has doubled every ~0.63 years for the past 6 years. At this pace, it is predicted that the structure of 90% of all human GPCRs will be completed within the next 5-7 years. This Review highlights the general structural features and principles that guide GPCR ligand recognition, receptor activation, G protein coupling, arrestin recruitment and regulation by GPCR kinases. The Review also highlights the diversity of GPCR allosteric binding sites and how allosteric ligands could dictate biased signalling that is selective for a G protein pathway or an arrestin pathway. Finally, the authors use the examples of glycoprotein hormone receptors and glucagon-like peptide 1 receptor to illustrate the effect of cryo-EM on understanding GPCR biology in endocrinology and metabolism, as well as on GPCR-related endocrine diseases and drug discovery.
Collapse
Affiliation(s)
- Jia Duan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Xin-Heng He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shu-Jie Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Department of Traditional Chinese Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
21
|
Parveen R, Hussain S, Saini S, Khan P, Saha N, Nidhi. Effect of ipragliflozin on liver enzymes in type 2 diabetes mellitus: a meta-analysis of randomized controlled trials. Expert Opin Pharmacother 2024; 25:925-935. [PMID: 38804904 DOI: 10.1080/14656566.2024.2360078] [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: 12/27/2023] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is estimated to affect upto 70-80% of people with type 2 diabetes mellitus (T2DM). Although several anti-hyperglycemic drugs have been shown to be effective in such patients, there remains an unmet need for newer drugs. The objective of this meta-analysis was to analyze the effect of ipragliflozin on aspartate aminotransferase (AST), alanine transaminase (ALT), and gamma-glutamyl transpeptidase (GGT) levels in patients with T2DM. METHODS A literature search on electronic databases was conducted to identify potential randomized clinical trials (RCT) as per predetermined study selection criteria. Mean difference (MD) was calculated using Cochrane review manager. RESULTS Twelve studies were included in the meta-analysis, including 1349 subjects. Compared to the control group, ipragliflozin as a monotherapy showed a significant reduction in levels of ALT at week 12 (p = 0.02) and at week 24 (p = 0.007), GGT at week 12 (p < 0.00001). Ipragliflozin as an add-on therapy showed significant reduction in levels of AST at week 24 (p < 0.00001), ALT at week 12 (p = 0.002), ALT at week 24 (p < 0.00001), and GGT at week 24 (p < 0.00001). CONCLUSION Findings suggest the beneficial effects of ipragliflozin on liver enzymes. Further large-scale RCTs are required to confirm ipragliflozin's role for liver-related conditions in T2DM.
Collapse
Affiliation(s)
- Rizwana Parveen
- Department of Translational and Clinical Research, Jamia Hamdard School of Chemical & Life Sciences, New Delhi, India
| | - Shadan Hussain
- Department of Translational and Clinical Research, Jamia Hamdard School of Chemical & Life Sciences, New Delhi, India
| | - Sparsh Saini
- Department of Translational and Clinical Research, Jamia Hamdard School of Chemical & Life Sciences, New Delhi, India
| | - Parvej Khan
- Department of Translational and Clinical Research, Jamia Hamdard School of Chemical & Life Sciences, New Delhi, India
| | - Nilanjan Saha
- Department of Translational and Clinical Research, Jamia Hamdard School of Chemical & Life Sciences, New Delhi, India
| | - Nidhi
- Department of Translational and Clinical Research, Jamia Hamdard School of Chemical & Life Sciences, New Delhi, India
| |
Collapse
|
22
|
Bavaresco A, Mazzeo P, Lazzara M, Barbot M. Adipose tissue in cortisol excess: What Cushing's syndrome can teach us? Biochem Pharmacol 2024; 223:116137. [PMID: 38494065 DOI: 10.1016/j.bcp.2024.116137] [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: 10/31/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Endogenous Cushing's syndrome (CS) is a rare condition due to prolonged exposure to elevated circulating cortisol levels that features its typical phenotype characterised by moon face, proximal myopathy, easy bruising, hirsutism in females and a centripetal distribution of body fat. Given the direct and indirect effects of hypercortisolism, CS is a severe disease burdened by increased cardio-metabolic morbidity and mortality in which visceral adiposity plays a leading role. Although not commonly found in clinical setting, endogenous CS is definitely underestimated leading to delayed diagnosis with consequent increased rate of complications and reduced likelihood of their reversal after disease control. Most of all, CS is a unique model for systemic impairment induced by exogenous glucocorticoid therapy that is commonly prescribed for a number of chronic conditions in a relevant proportion of the worldwide population. In this review we aim to summarise on one side, the mechanisms behind visceral adiposity and lipid metabolism impairment in CS during active disease and after remission and on the other explore the potential role of cortisol in promoting adipose tissue accumulation.
Collapse
Affiliation(s)
- Alessandro Bavaresco
- Department of Medicine DIMED, University of Padua, Padua, Italy; Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padua, Padua, Italy
| | - Pierluigi Mazzeo
- Department of Medicine DIMED, University of Padua, Padua, Italy; Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padua, Padua, Italy
| | - Martina Lazzara
- Department of Medicine DIMED, University of Padua, Padua, Italy; Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padua, Padua, Italy
| | - Mattia Barbot
- Department of Medicine DIMED, University of Padua, Padua, Italy; Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padua, Padua, Italy.
| |
Collapse
|
23
|
Nandi A, Zhang A, Arad E, Jelinek R, Warshel A. Assessing the Catalytic Role of Native Glucagon Amyloid Fibrils. ACS Catal 2024; 14:4656-4664. [PMID: 39070231 PMCID: PMC11270920 DOI: 10.1021/acscatal.4c00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Glucagon stands out as a pivotal peptide hormone, instrumental in controlling blood glucose levels and lipid metabolism. While the formation of glucagon amyloid fibrils has been documented, their biological functions remain enigmatic. Recently, we demonstrated experimentally that glucagon amyloid fibrils can act as catalysts in several biological reactions including esterolysis, lipid hydrolysis, and dephosphorylation. Herein, we present a multiscale quantum mechanics/molecular mechanics (QM/MM) simulation of the acylation step in the esterolysis of para-nitrophenyl acetate (p-NPA), catalyzed by native glucagon amyloid fibrils, serving as a model system to elucidate their catalytic function. This step entails a concerted mechanism, involving proton transfer from serine to histidine, followed by the nucleophilic attack of the serine oxy anion on the carbonyl carbon of p-NPA. We computed the binding energy and free-energy profiles of this reaction using the protein-dipole Langevin-dipole (PDLD) within the linear response approximation (LRA) framework (PDLD/S-LRA-2000) and the empirical valence bond (EVB) methods. This included simulations of the reaction in an aqueous environment and in the fibril, enabling us to estimate the catalytic effect of the fibril. Our EVB calculations obtained a barrier of 23.4 kcal mol-1 for the enzyme-catalyzed reaction compared to the experimental value of 21.9 kcal mol-1 (and a calculated catalytic effect of 3.2 kcal mol-1 compared to the observed effect of 4.7 kcal mol-1). This close agreement together with the barrier reduction when transitioning from the reference solution reaction to the amyloid fibril provides supporting evidence to the catalytic role of glucagon amyloid fibrils. Moreover, employing the PDLD/S-LRA-2000 approach further reinforced exclusively the enzyme's catalytic role. The results presented in this study contribute significantly to our understanding of the catalytic role of glucagon amyloid fibrils, marking, to the best of our knowledge, the first-principles mechanistic investigation of fibrils using QM/MM methods. Therefore, our findings offer fruitful insights for future research into the mechanisms of related amyloid catalysis.
Collapse
Affiliation(s)
- Ashim Nandi
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| | - Aoxuan Zhang
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| | - Elad Arad
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Chemical Engineering, Columbia University, New York, New York 10027, United States
| | - Raz Jelinek
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| |
Collapse
|
24
|
Hope DCD, Ansari S, Choudhury S, Alexiadou K, Tabbakh Y, Ilesanmi I, Lazarus K, Davies I, Jimenez-Pacheco L, Yang W, Ball LJ, Malviya R, Reglinska B, Khoo B, Minnion J, Bloom SR, Tan TMM. Adaptive infusion of a glucagon-like peptide-1/glucagon receptor co-agonist G3215, in adults with overweight or obesity: Results from a phase 1 randomized clinical trial. Diabetes Obes Metab 2024; 26:1479-1491. [PMID: 38229453 DOI: 10.1111/dom.15448] [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: 11/17/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024]
Abstract
AIMS To determine whether a continuous infusion of a glucagon-like peptide receptor (GLP-1R)/glucagon receptor (GCGR) co-agonist, G3215 is safe and well tolerated in adults with overweight or obesity. METHODS A phase 1 randomized, double blind, placebo-controlled trial of G3215 in overweight or obese participants, with or without type 2 diabetes. RESULTS Twenty-six participants were recruited and randomized with 23 completing a 14-day subcutaneous infusion of G3215 or placebo. The most common adverse events were nausea or vomiting, which were mild in most cases and mitigated by real-time adjustment of drug infusion. There were no cardiovascular concerns with G3215 infusion. The pharmacokinetic characteristics were in keeping with a continuous infusion over 14 days. A least-squares mean body weight loss of 2.39 kg was achieved with a 14-day infusion of G3215, compared with 0.84 kg with placebo infusion (p < .05). A reduction in food consumption was also observed in participants receiving G3215 and there was no deterioration in glycaemia. An improved lipid profile was seen in G3215-treated participants and consistent with GCGR activation, a broad reduction in circulating amino acids was seen during the infusion period. CONCLUSION An adaptive continuous infusion of the GLP-1/GCGR co-agonist, G3215, is safe and well tolerated offering a unique strategy to control drug exposure. By allowing rapid, response-directed titration, this strategy may allow for mitigation of adverse effects and afford significant weight loss within shorter time horizons than is presently possible with weekly GLP-1R and multi-agonists. These results support ongoing development of G3215 for the treatment of obesity and metabolic disease.
Collapse
Affiliation(s)
- David C D Hope
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Saleem Ansari
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Sirazum Choudhury
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Kleopatra Alexiadou
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Yasmin Tabbakh
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ibiyemi Ilesanmi
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Katharine Lazarus
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Iona Davies
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Lara Jimenez-Pacheco
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Wei Yang
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Laura-Jayne Ball
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Reshma Malviya
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Beata Reglinska
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Bernard Khoo
- Endocrinology, Division of Medicine, University College London, London, UK
| | - James Minnion
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stephen R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M-M Tan
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
25
|
Chrysavgis LG, Kazanas S, Bafa K, Rozani S, Koloutsou ME, Cholongitas E. Glucagon-like Peptide 1, Glucose-Dependent Insulinotropic Polypeptide, and Glucagon Receptor Agonists in Metabolic Dysfunction-Associated Steatotic Liver Disease: Novel Medication in New Liver Disease Nomenclature. Int J Mol Sci 2024; 25:3832. [PMID: 38612640 PMCID: PMC11012092 DOI: 10.3390/ijms25073832] [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: 02/11/2024] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are incretins that regulate postprandial glucose regulation, stimulating insulin secretion from pancreatic β-cells in response to food ingestion. Modified GLP-1 receptor agonists (GLP-1RAs) are being administered for the treatment of obesity and type 2 diabetes mellitus (T2DM). Strongly related to those disorders, metabolic dysfunction-associated steatotic liver disease (MASLD), especially its aggressive form, defined as metabolic dysfunction-associated steatohepatitis (MASH), is a major healthcare burden associated with high morbidity and extrahepatic complications. GLP-1RAs have been explored in MASH patients with evident improvement in liver dysfunction enzymes, glycemic control, and weight loss. Importantly, the combination of GLP-1RAs with GIP and/or glucagon RAs may be even more effective via synergistic mechanisms in amelioration of metabolic, biochemical, and histological parameters of MASLD but also has a beneficial impact on MASLD-related complications. In this current review, we aim to provide an overview of incretins' physiology, action, and signaling. Furthermore, we provide insight into the key pathophysiological mechanisms through which they impact MASLD aspects, as well as we analyze clinical data from human interventional studies. Finally, we discuss the current challenges and future perspectives pertinent to this growing area of research and clinical medicine.
Collapse
Affiliation(s)
- Lampros G. Chrysavgis
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, General Hospital Laiko, 115 27 Athens, Greece; (L.G.C.); (S.K.); (K.B.); (S.R.)
| | - Spyridon Kazanas
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, General Hospital Laiko, 115 27 Athens, Greece; (L.G.C.); (S.K.); (K.B.); (S.R.)
| | - Konstantina Bafa
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, General Hospital Laiko, 115 27 Athens, Greece; (L.G.C.); (S.K.); (K.B.); (S.R.)
| | - Sophia Rozani
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, General Hospital Laiko, 115 27 Athens, Greece; (L.G.C.); (S.K.); (K.B.); (S.R.)
| | - Maria-Evangelia Koloutsou
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, General Hospital Laiko, 115 27 Athens, Greece;
| | - Evangelos Cholongitas
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, General Hospital Laiko, 115 27 Athens, Greece; (L.G.C.); (S.K.); (K.B.); (S.R.)
| |
Collapse
|
26
|
Wang MY, Zhang Z, Zhao S, Onodera T, Sun XN, Zhu Q, Li C, Li N, Chen S, Paredes M, Gautron L, Charron MJ, Marciano DK, Gordillo R, Drucker DJ, Scherer PE. Downregulation of the kidney glucagon receptor, essential for renal function and systemic homeostasis, contributes to chronic kidney disease. Cell Metab 2024; 36:575-597.e7. [PMID: 38237602 PMCID: PMC10932880 DOI: 10.1016/j.cmet.2023.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 09/10/2023] [Accepted: 12/19/2023] [Indexed: 02/12/2024]
Abstract
The glucagon receptor (GCGR) in the kidney is expressed in nephron tubules. In humans and animal models with chronic kidney disease, renal GCGR expression is reduced. However, the role of kidney GCGR in normal renal function and in disease development has not been addressed. Here, we examined its role by analyzing mice with constitutive or conditional kidney-specific loss of the Gcgr. Adult renal Gcgr knockout mice exhibit metabolic dysregulation and a functional impairment of the kidneys. These mice exhibit hyperaminoacidemia associated with reduced kidney glucose output, oxidative stress, enhanced inflammasome activity, and excess lipid accumulation in the kidney. Upon a lipid challenge, they display maladaptive responses with acute hypertriglyceridemia and chronic proinflammatory and profibrotic activation. In aged mice, kidney Gcgr ablation elicits widespread renal deposition of collagen and fibronectin, indicative of fibrosis. Taken together, our findings demonstrate an essential role of the renal GCGR in normal kidney metabolic and homeostatic functions. Importantly, mice deficient for kidney Gcgr recapitulate some of the key pathophysiological features of chronic kidney disease.
Collapse
Affiliation(s)
- May-Yun Wang
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shangang Zhao
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Sam and Ann Barshop Institute for Longevity and Aging Studies, Division of Endocrinology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Toshiharu Onodera
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xue-Nan Sun
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qingzhang Zhu
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chao Li
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Na Li
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Megan Paredes
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laurent Gautron
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Denise K Marciano
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel J Drucker
- Lunenfeld-TanenbaumResearchInstitute, Mt. Sinai Hospital, Toronto, ON M5G1X5, Canada; Department of Medicine, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
27
|
Pešić D, Đukić MM, Stanojević I, Živkovć V, Bolevich S, Bolevich S, Jakovljević V. Cardiorespiratory fitness mediates cortisol and lactate responses to winter and summer marches. J Med Biochem 2024; 43:72-85. [PMID: 38496029 PMCID: PMC10943469 DOI: 10.5937/jomb0-44369] [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: 05/15/2023] [Accepted: 06/26/2023] [Indexed: 03/19/2024] Open
Abstract
Background The influence of homeostatically regulated physiological processes, including cardiorespiratory fitness (VO2max), on the response to physical stressors such as acclimatisation and marching, remains understudied. We aimed to investigate the effects of summer and winter acclimatisation and marching on cortisol levels and blood lactate, to gain insight into the role of these physiological processes in the stress response. Methods Two groups of young Europeans, classified as poor (PCF; n=9) and good physical condition (GCF; n=21), based on a VO2MAX threshold of 40 mL O2/ kg/min, underwent 2-h March (6-7 km/h) in winter (5˚C) and summer (32˚C). Commercial tests, UniCel DxI Access Cortisol assay and EKF Biosen Clinic/GP assay were used for cortisol and lactate blood measurements (morning samples and those taken immediately after marches), respectively.
Collapse
Affiliation(s)
- Deniel Pešić
- Military Medical Academy, Institute of Hygiene, Department of Exercise Physiology, Belgrade
| | - Mirjana M. Đukić
- University of Belgrade, Faculty of Pharmacy, Department of Toxicology, Belgrade
| | - Ivan Stanojević
- Military Medical Academy, Institute of Medical Research, Belgrade
| | - Vladimir Živkovć
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Kragujevac
| | - Sergey Bolevich
- First Moscow State Medical University I. M. Sechenov, Department of Pharmacology, Moscow, Russia
| | - Stefani Bolevich
- First Moscow State Medical University I. M. Sechenov, Department of Pharmacology, Moscow, Russia
| | - Vladimir Jakovljević
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Kragujevac
| |
Collapse
|
28
|
Asadi F, Gunawardana SC, Dolle RE, Piston DW. An orally available compound suppresses glucagon hypersecretion and normalizes hyperglycemia in type 1 diabetes. JCI Insight 2024; 9:e172626. [PMID: 38258903 PMCID: PMC10906223 DOI: 10.1172/jci.insight.172626] [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: 05/31/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Suppression of glucagon hypersecretion can normalize hyperglycemia during type 1 diabetes (T1D). Activating erythropoietin-producing human hepatocellular receptor type-A4 (EphA4) on α cells reduced glucagon hypersecretion from dispersed α cells and T1D islets from both human donor and mouse models. We synthesized a high-affinity small molecule agonist for the EphA4 receptor, WCDD301, which showed robust plasma and liver microsome metabolic stability in both mouse and human preparations. In islets and dispersed islet cells from nondiabetic and T1D human donors, WCDD301 reduced glucagon secretion comparable to the natural EphA4 ligand, Ephrin-A5. In diabetic NOD and streptozotocin-treated mice, once-daily oral administration of WCDD301 formulated with a time-release excipient reduced plasma glucagon and normalized blood glucose for more than 3 months. These results suggest that targeting the α cell EphA4 receptor by sustained release of WCDD301 is a promising pharmacologic pathway for normalizing hyperglycemia in patients with T1D.
Collapse
Affiliation(s)
| | | | - Roland E. Dolle
- Center for Drug Discovery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
29
|
Klein T, Augustin R, Hennige AM. Perspectives in weight control in diabetes - Survodutide. Diabetes Res Clin Pract 2024; 207:110779. [PMID: 37330144 DOI: 10.1016/j.diabres.2023.110779] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/19/2023]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists are approved treatments for Type 2 diabetes mellitus, with liraglutide and semaglutide also approved for the treatment of obesity. The natural gut hormone oxyntomodulin is a weak dual agonist of the glucagon receptor (GCGR) and GLP-1R. Development of poly-agonists mimicking oxyntomodulin, such as the novel dual GCGR/GLP-1R agonist survodutide, represents an important step towards a more effective treatment for people with Type 2 diabetes mellitus and obesity. Survodutide is a 29-amino acid peptide derived from glucagon, with the incorporation of potent GLP-1 activities. It contains a C18 diacid which mediates binding to albumin, thereby prolonging the half-life to enable once-weekly subcutaneous dosing. The utilisation of GCGR agonism aims to enhance body weight-lowering effects by increasing energy expenditure in addition to the anorectic action of GLP-1R agonists. Glucose-lowering efficacy of survodutide has been demonstrated in a Phase II trial in patients with Type 2 diabetes mellitus and obesity and was associated with clinically meaningful body weight loss. These data highlight the potential of dual GCGR/GLP-1R agonism for reducing glycated haemoglobin and body weight in patients with Type 2 diabetes mellitus, and for greater therapeutic efficacy compared with GLP-1R agonism alone.
Collapse
Affiliation(s)
- Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88397, Biberach an der Riss, Germany
| | - Robert Augustin
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88397, Biberach an der Riss, Germany
| | - Anita M Hennige
- Boehringer Ingelheim International GmbH, Birkendorfer Strasse 65, 88397, Biberach an der Riss, Germany.
| |
Collapse
|
30
|
Egners A, Cramer T, Wallach I, Berndt N. Kinetic Modeling of Hepatic Metabolism and Simulation of Treatment Effects. Methods Mol Biol 2024; 2769:211-225. [PMID: 38315400 DOI: 10.1007/978-1-0716-3694-7_16] [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] [Indexed: 02/07/2024]
Abstract
Mathematical modeling is a promising strategy to fill the experimentally unapproachable knowledge gaps about the relative contribution of various molecular processes to cellular metabolic function. To this end, we developed detailed kinetic models of the central metabolism of different cell types, comprising multiple metabolic functionalities. We used the model to simulate metabolic changes in several cell types under different experimental settings in health and disease. In this way, we show that it is possible to decipher and characterize the relative influence of various metabolic pathways and enzymes to overall metabolic performance and phenotype.Quantitative Systems Metabolism (QSM™) allows quantitative assessment of metabolic functionality and metabolic profiling based on proteomic data. Here, we describe the technique, namely, molecular resolved kinetic modeling, underlying QSM™. We explain the necessary steps for the generation of cell-specific models to functionally interpret proteomic data and point out some unresolved challenges and open questions.
Collapse
Affiliation(s)
- Antje Egners
- Molecular Tumor Biology, Department of General, Visceral and Transplantation Surgery, RWTH University Hospital, Aachen, Germany
| | - Thorsten Cramer
- Molecular Tumor Biology, Department of General, Visceral and Transplantation Surgery, RWTH University Hospital, Aachen, Germany
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Iwona Wallach
- Deutsches Herzzentrum der Charité (DHZC), Institute of Computer-assisted Cardiovascular Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nikolaus Berndt
- Deutsches Herzzentrum der Charité (DHZC), Institute of Computer-assisted Cardiovascular Medicine, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany.
| |
Collapse
|
31
|
Lim JJ, Sequeira-Bisson IR, Yip WCY, Lu LW, Miles-Chan JL, Poppitt SD. Intra-pancreatic fat is associated with high circulating glucagon and GLP-1 concentrations following whey protein ingestion in overweight women with impaired fasting glucose: A randomised controlled trial. Diabetes Res Clin Pract 2024; 207:111084. [PMID: 38154534 DOI: 10.1016/j.diabres.2023.111084] [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: 09/06/2023] [Revised: 12/13/2023] [Accepted: 12/25/2023] [Indexed: 12/30/2023]
Abstract
AIM Intra-pancreatic fat deposition (IPFD) while hypothesised to impair beta-cell function, its impact on alpha-cells remains unclear. We evaluated the association between IPFD and markers of pancreatic cells function using whey protein. METHODS Twenty overweight women with impaired fasting glucose (IFG) and low or high IPFD (<4.66% vs ≥4.66%) consumed 3 beverage treatments: 0 g (water control), 12.5 g (low-dose) and 50.0 g (high-dose) whey protein, after an overnight fast, in randomised order. Blood glucose, insulin, C-peptide, glucagon, gastric-inhibitory polypeptide (GIP), glucagon-like peptide-1 (GLP-1) and amylin were analysed postprandially over 4 h. Incremental area-under-the-curve (iAUC), incremental maximum concentration (iCmax), and time to maximum concentration (Tmax) for these were compared between IPFD groups using repeated measures linear mixed models, also controlled for age (pcov). RESULTS iAUC and iCmax glucose and insulin while similar between the two IPFD groups, high IPFD and ageing contributed to higher postprandial glucagon (iAUC: p = 0.012; pcov = 0.004; iCmax: p = 0.069; pcov = 0.021) and GLP-1 (iAUC: p = 0.006; pcov = 0.064; iCmax: p = 0.011; pcov = 0.122) concentrations. CONCLUSION In our cohort, there was no evidence that IPFD impaired protein-induced insulin secretion. Conversely, IPFD may be associated with increased protein-induced glucagon secretion, a novel observation which warrants further investigation into its relevance in the pathogenesis of dysglycaemia and type-2 diabetes.
Collapse
Affiliation(s)
- Jia Jiet Lim
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand; Riddet Institute, Palmerston North, New Zealand; High Value Nutrition, National Science Challenge, Auckland, New Zealand.
| | - Ivana R Sequeira-Bisson
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand; Riddet Institute, Palmerston North, New Zealand; High Value Nutrition, National Science Challenge, Auckland, New Zealand
| | - Wilson C Y Yip
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand; High Value Nutrition, National Science Challenge, Auckland, New Zealand
| | - Louise W Lu
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand; High Value Nutrition, National Science Challenge, Auckland, New Zealand
| | - Jennifer L Miles-Chan
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand; Riddet Institute, Palmerston North, New Zealand; High Value Nutrition, National Science Challenge, Auckland, New Zealand
| | - Sally D Poppitt
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand; Riddet Institute, Palmerston North, New Zealand; High Value Nutrition, National Science Challenge, Auckland, New Zealand; Department of Medicine, University of Auckland, Auckland, New Zealand
| |
Collapse
|
32
|
Ang T, Mason SA, Dao GM, Bruce CR, Kowalski GM. The impact of a single dose of whey protein on glucose flux and metabolite profiles in normoglycemic males: insights into glucagon and insulin biology. Am J Physiol Endocrinol Metab 2023; 325:E688-E699. [PMID: 37877796 DOI: 10.1152/ajpendo.00182.2023] [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: 06/20/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023]
Abstract
Protein ingestion concurrently stimulates euglycemic glucagon and insulin secretion, a response that is particularly robust with rapidly absorbing proteins. Previously, we have shown that ingestion of repeated doses of rapidly absorbing whey protein equally stimulated endogenous glucose production (EGP) and glucose disposal (Rd), thus explaining the preservation of euglycemia. Here, we aimed to determine if a smaller single dose of whey could elicit a large enough glucagon and insulin response to stimulate glucose flux. Therefore, in normoglycemic young adult males (n = 10; age ∼26; BMI ∼25), using [6,6-2H2] glucose tracing and quantitative targeted metabolite profiling, we determined the metabolic response to a single 25 g "standard" dose of whey protein. Whey protein ingestion did not alter glycemia, but increased circulating glucagon (peak 4-fold basal), insulin (peak 6-fold basal), amino acids, and urea while also reducing free fatty acid (FFA) and glycerol concentrations. Interestingly, the postprandial insulin response was driven by both a stimulation of insulin secretion and marked reduction in hepatic insulin clearance. Whey protein ingestion resulted in a modest stimulation of EGP and Rd, both peaking at ∼20% above baseline 1 h after protein ingestion. These findings demonstrate that the ingestion of a single standard serving of whey protein can induce a euglycemic glucagon and insulin response that stimulates glucose flux. We speculate on a theory that could potentially explain how glucagon and insulin synergistically provide hardwired control of nitrogen and glucose homeostasis.NEW & NOTEWORTHY Protein ingestion concurrently stimulates glucagon and insulin secretion. Here we show that in normoglycemic males, ingestion of a single "standard" 25 g serving of rapidly absorbing whey protein drives a sufficiently large glucagon and insulin response, such that it simultaneously increases endogenous glucose production and glucose disposal. We speculate on a novel theory that could potentially explain how the antagonistic/synergistic actions of glucagon and insulin simultaneously provide tight control of glucose and nitrogen homeostasis.
Collapse
Affiliation(s)
- Teddy Ang
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
- School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
| | - Shaun A Mason
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
- School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
| | - Giang M Dao
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Clinton R Bruce
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
- School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
| | - Greg M Kowalski
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Geelong, Victoria, Australia
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
33
|
Peng J, Yu L, Huang L, Paschoal VA, Chu H, de Souza CO, Varre JV, Oh DY, Kohler JJ, Xiao X, Xu L, Holland WL, Shaul PW, Mineo C. Hepatic sialic acid synthesis modulates glucose homeostasis in both liver and skeletal muscle. Mol Metab 2023; 78:101812. [PMID: 37777009 PMCID: PMC10583174 DOI: 10.1016/j.molmet.2023.101812] [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: 08/01/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
OBJECTIVE Sialic acid is a terminal monosaccharide of glycans in glycoproteins and glycolipids, and its derivation from glucose is regulated by the rate-limiting enzyme UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE). Although the glycans on key endogenous hepatic proteins governing glucose metabolism are sialylated, how sialic acid synthesis and sialylation in the liver influence glucose homeostasis is unknown. Studies were designed to fill this knowledge gap. METHODS To decrease the production of sialic acid and sialylation in hepatocytes, a hepatocyte-specific GNE knockdown mouse model was generated, and systemic glucose metabolism, hepatic insulin signaling and glucagon signaling were evaluated in vivo or in primary hepatocytes. Peripheral insulin sensitivity was also assessed. Furthermore, the mechanisms by which sialylation in the liver influences hepatic insulin signaling and glucagon signaling and peripheral insulin sensitivity were identified. RESULTS Liver GNE deletion in mice caused an impairment of insulin suppression of hepatic glucose production. This was due to a decrease in the sialylation of hepatic insulin receptors (IR) and a decline in IR abundance due to exaggerated degradation through the Eph receptor B4. Hepatic GNE deficiency also caused a blunting of hepatic glucagon receptor (GCGR) function which was related to a decline in its sialylation and affinity for glucagon. An accompanying upregulation of hepatic FGF21 production caused an enhancement of skeletal muscle glucose disposal that led to an overall increase in glucose tolerance and insulin sensitivity. CONCLUSION These collective observations reveal that hepatic sialic acid synthesis and sialylation modulate glucose homeostasis in both the liver and skeletal muscle. By interrogating how hepatic sialic acid synthesis influences glucose control mechanisms in the liver, a new metabolic cycle has been identified in which a key constituent of glycans generated from glucose modulates the systemic control of its precursor.
Collapse
Affiliation(s)
- Jun Peng
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Liming Yu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Linzhang Huang
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Vivian A Paschoal
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Haiyan Chu
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Camila O de Souza
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Joseph V Varre
- Dept. of Nutrition & Integrative Physiology, University of Utah College of Health, 250 1850 E, Salt Lake City, UT, 84112, USA
| | - Da Young Oh
- Dept. of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Jennifer J Kohler
- Dept. of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xue Xiao
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Lin Xu
- Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - William L Holland
- Dept. of Nutrition & Integrative Physiology, University of Utah College of Health, 250 1850 E, Salt Lake City, UT, 84112, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Dept. of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA; Dept. of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| |
Collapse
|
34
|
Kim J, Chang N, Kim Y, Lee J, Oh D, Choi J, Kim O, Kim S, Choi M, Lee J, Lee J, Kim J, Cho M, Kim M, Lee K, Hwang D, Sa JK, Park S, Baek S, Im D. The Novel Tetra-Specific Drug C-192, Conjugated Using UniStac, Alleviates Non-Alcoholic Steatohepatitis in an MCD Diet-Induced Mouse Model. Pharmaceuticals (Basel) 2023; 16:1601. [PMID: 38004466 PMCID: PMC10674394 DOI: 10.3390/ph16111601] [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: 09/11/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a complex disease resulting from chronic liver injury associated with obesity, type 2 diabetes, and inflammation. Recently, the importance of developing multi-target drugs as a strategy to address complex diseases such as NASH has been growing; however, their manufacturing processes remain time- and cost-intensive and inefficient. To overcome these limitations, we developed UniStac, a novel enzyme-mediated conjugation platform for multi-specific drug development. UniStac demonstrated high conjugation yields, optimal thermal stabilities, and robust biological activities. We designed a tetra-specific compound, C-192, targeting glucagon-like peptide 1 (GLP-1), glucagon (GCG), fibroblast growth factor 21 (FGF21), and interleukin-1 receptor antagonist (IL-1RA) simultaneously for the treatment of NASH using UniStac. The biological activity and treatment efficacy of C-192 were confirmed both in vitro and in vivo using a methionine-choline-deficient (MCD) diet-induced mouse model. C-192 exhibited profound therapeutic efficacies compared to conventional drugs, including liraglutide and dulaglutide. C-192 significantly improved alanine transaminase levels, triglyceride accumulation, and the non-alcoholic fatty liver disease activity score. In this study, we demonstrated the feasibility of UniStac in creating multi-specific drugs and confirmed the therapeutic potential of C-192, a drug that integrates multiple mechanisms into a single molecule for the treatment of NASH.
Collapse
Affiliation(s)
- Jihye Kim
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Nakho Chang
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Yunki Kim
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Jaehyun Lee
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Daeseok Oh
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Jaeyoung Choi
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Onyou Kim
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Sujin Kim
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Myongho Choi
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Junyeob Lee
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Junghwa Lee
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Jungyul Kim
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Minji Cho
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Minsu Kim
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Kwanghwan Lee
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Dukhyun Hwang
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Jason K. Sa
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Sungjin Park
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| | - Seungjae Baek
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Daeseong Im
- Onegene Biotechnology, Inc., 205 Ace Gwanggyo Tower 2, 91 Changnyong-daero 256 beon-gil, Yeongtong-gu, Suwon-si 16229, Republic of Korea; (J.K.); (J.C.); (J.K.); (K.L.)
| |
Collapse
|
35
|
Genchi VA, Palma G, Sorice GP, D'Oria R, Caccioppoli C, Marrano N, Biondi G, Caruso I, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Pharmacological modulation of adaptive thermogenesis: new clues for obesity management? J Endocrinol Invest 2023; 46:2213-2236. [PMID: 37378828 PMCID: PMC10558388 DOI: 10.1007/s40618-023-02125-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Adaptive thermogenesis represents the main mechanism through which the body generates heat in response to external stimuli, a phenomenon that includes shivering and non-shivering thermogenesis. The non-shivering thermogenesis is mainly exploited by adipose tissue characterized by a brown aspect, which specializes in energy dissipation. A decreased amount of brown adipose tissue has been observed in ageing and chronic illnesses such as obesity, a worldwide health problem characterized by dysfunctional adipose tissue expansion and associated cardiometabolic complications. In the last decades, the discovery of a trans-differentiation mechanism ("browning") within white adipose tissue depots, leading to the generation of brown-like cells, allowed to explore new natural and synthetic compounds able to favour this process and thus enhance thermogenesis with the aim of counteracting obesity. Based on recent findings, brown adipose tissue-activating agents could represent another option in addition to appetite inhibitors and inhibitors of nutrient absorption for obesity treatment. PURPOSE This review investigates the main molecules involved in the physiological (e.g. incretin hormones) and pharmacological (e.g. β3-adrenergic receptors agonists, thyroid receptor agonists, farnesoid X receptor agonists, glucagon-like peptide-1, and glucagon receptor agonists) modulation of adaptive thermogenesis and the signalling mechanisms involved.
Collapse
Affiliation(s)
- V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G Palma
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G P Sorice
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - R D'Oria
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - C Caccioppoli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - N Marrano
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G Biondi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - I Caruso
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
36
|
Zhang J, Zheng Y, Martens L, Pfeiffer AFH. The Regulation and Secretion of Glucagon in Response to Nutrient Composition: Unraveling Their Intricate Mechanisms. Nutrients 2023; 15:3913. [PMID: 37764697 PMCID: PMC10536047 DOI: 10.3390/nu15183913] [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: 08/01/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Glucagon was initially regarded as a hyperglycemic substance; however, recent research has revealed its broader role in metabolism, encompassing effects on glucose, amino acids (AAs), and lipid metabolism. Notably, the interplay of glucagon with nutrient intake, particularly of AAs, and non-nutrient components is central to its secretion. Fasting and postprandial hyperglucagonemia have long been linked to the development and progression of type 2 diabetes (T2DM). However, recent studies have brought to light the positive impact of glucagon agonists on lipid metabolism and energy homeostasis. This review explores the multifaceted actions of glucagon, focusing on its regulation, signaling pathways, and effects on glucose, AAs, and lipid metabolism. The interplay between glucagon and other hormones, including insulin and incretins, is examined to provide a mechanistic understanding of its functions. Notably, the liver-α-cell axis, which involves glucagon and amino acids, emerges as a critical aspect of metabolic regulation. The dysregulation of glucagon secretion and its impact on conditions such as T2DM are discussed. The review highlights the potential therapeutic applications of targeting the glucagon pathway in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Jiudan Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China;
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
| | - Yang Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Lisa Martens
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
- Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
| |
Collapse
|
37
|
McGlone ER, Siebert M, Dore M, Hope DCD, Davies I, Owen B, Khoo B, Goldin R, Carling D, Bloom S, Le Gall M, Tan TM. Sleeve gastrectomy causes weight-loss independent improvements in hepatic steatosis. Liver Int 2023; 43:1890-1900. [PMID: 37208943 PMCID: PMC10947097 DOI: 10.1111/liv.15614] [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: 09/14/2022] [Revised: 03/15/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND AND AIMS Sleeve gastrectomy (VSG) leads to improvement in hepatic steatosis, associated with weight loss. The aims of this study were to investigate whether VSG leads to weight-loss independent improvements in liver steatosis in mice with diet-induced obesity (DIO); and to metabolically and transcriptomically profile hepatic changes in mice undergoing VSG. METHODS Mice with DIO were treated with VSG, sham surgery with subsequent food restriction to weight-match to the VSG group (Sham-WM), or sham surgery with return to unrestricted diet (Sham-Ad lib). Hepatic steatosis, glucose tolerance, insulin and glucagon resistance, and hepatic transcriptomics were investigated at the end of the study period and treatment groups were compared with mice undergoing sham surgery only (Sham-Ad lib). RESULTS VSG led to much greater improvement in liver steatosis than Sham-WM (liver triglyceride mg/mg 2.5 ± 0.1, 2.1 ± 0.2, 1.6 ± 0.1 for Sham-AL, Sham-WM and VSG respectively; p = 0.003). Homeostatic model assessment of insulin resistance was improved following VSG only (51.2 ± 8.8, 36.3 ± 5.3, 22.3 ± 6.1 for Sham-AL, Sham-WM and VSG respectively; p = 0.03). The glucagon-alanine index, a measure of glucagon resistance, fell with VSG but was significantly increased in Sham-WM (9.8 ± 1.7, 25.8 ± 4.6 and 5.2 ± 1.2 in Sham Ad-lib, Sham-WM and VSG respectively; p = 0.0003). Genes downstream of glucagon receptor signalling which govern fatty acid synthesis (Acaca, Acacb, Me1, Acly, Fasn and Elovl6) were downregulated following VSG but upregulated in Sham-WM. CONCLUSIONS Changes in glucagon sensitivity may contribute to weight-loss independent improvements in hepatic steatosis following VSG.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Department of Surgery and CancerImperial College LondonLondonUK
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Matthieu Siebert
- Centre de Recherche sur l'Inflammation, UMRS1149, Inserm, Université Paris CitéParisFrance
| | - Marian Dore
- Genomics FacilityMRC London Institute of Medical Sciences (LMS), Imperial College LondonLondonUK
| | - David C. D. Hope
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Iona Davies
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Bryn Owen
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Bernard Khoo
- Division of MedicineUniversity College London, Royal Free HospitalLondonUK
| | - Rob Goldin
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Dave Carling
- Cellular Stress GroupMRC LMS, Imperial College LondonLondonUK
| | - Stephen Bloom
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Maude Le Gall
- Centre de Recherche sur l'Inflammation, UMRS1149, Inserm, Université Paris CitéParisFrance
| | - Tricia M‐M. Tan
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| |
Collapse
|
38
|
Armour SL, Stanley JE, Cantley J, Dean ED, Knudsen JG. Metabolic regulation of glucagon secretion. J Endocrinol 2023; 259:e230081. [PMID: 37523232 PMCID: PMC10681275 DOI: 10.1530/joe-23-0081] [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: 03/17/2023] [Accepted: 07/31/2023] [Indexed: 08/01/2023]
Abstract
Since the discovery of glucagon 100 years ago, the hormone and the pancreatic islet alpha cells that produce it have remained enigmatic relative to insulin-producing beta cells. Canonically, alpha cells have been described in the context of glucagon's role in glucose metabolism in liver, with glucose as the primary nutrient signal regulating alpha cell function. However, current data reveal a more holistic model of metabolic signalling, involving glucagon-regulated metabolism of multiple nutrients by the liver and other tissues, including amino acids and lipids, providing reciprocal feedback to regulate glucagon secretion and even alpha cell mass. Here we describe how various nutrients are sensed, transported and metabolised in alpha cells, providing an integrative model for the metabolic regulation of glucagon secretion and action. Importantly, we discuss where these nutrient-sensing pathways intersect to regulate alpha cell function and highlight key areas for future research.
Collapse
Affiliation(s)
- Sarah L Armour
- Section for cell biology and physiology, Department of Biology, University of Copenhagen, DK
| | - Jade E. Stanley
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, USA
| | - James Cantley
- Division of Cellular and systems medicine, School of Medicine, University of Dundee, UK
| | - E. Danielle Dean
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, USA
- Division of Diabetes, Endocrinology, & Metabolism, Vanderbilt University Medical Center school of medicine, USA
| | - Jakob G Knudsen
- Section for cell biology and physiology, Department of Biology, University of Copenhagen, DK
| |
Collapse
|
39
|
Dagdeviren S, Hoang MF, Sarikhani M, Meier V, Benoit JC, Okawa MC, Melnik VY, Ricci-Blair EM, Foot N, Friedline RH, Hu X, Tauer LA, Srinivasan A, Prigozhin MB, Shenoy SK, Kumar S, Kim JK, Lee RT. An insulin-regulated arrestin domain protein controls hepatic glucagon action. J Biol Chem 2023; 299:105045. [PMID: 37451484 PMCID: PMC10413355 DOI: 10.1016/j.jbc.2023.105045] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/16/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
Glucagon signaling is essential for maintaining normoglycemia in mammals. The arrestin fold superfamily of proteins controls the trafficking, turnover, and signaling of transmembrane receptors as well as other intracellular signaling functions. Further investigation is needed to understand the in vivo functions of the arrestin domain-containing 4 (ARRDC4) protein family member and whether it is involved in mammalian glucose metabolism. Here, we show that mice with a global deletion of the ARRDC4 protein have impaired glucagon responses and gluconeogenesis at a systemic and molecular level. Mice lacking ARRDC4 exhibited lower glucose levels after fasting and could not suppress gluconeogenesis at the refed state. We also show that ARRDC4 coimmunoprecipitates with the glucagon receptor, and ARRDC4 expression is suppressed by insulin. These results define ARRDC4 as a critical regulator of glucagon signaling and glucose homeostasis and reveal a novel intersection of insulin and glucagon pathways in the liver.
Collapse
Affiliation(s)
- Sezin Dagdeviren
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Megan F Hoang
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Mohsen Sarikhani
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Vanessa Meier
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Jake C Benoit
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Marinna C Okawa
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Veronika Y Melnik
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Elisabeth M Ricci-Blair
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Natalie Foot
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Randall H Friedline
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Xiaodi Hu
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Lauren A Tauer
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Arvind Srinivasan
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Maxim B Prigozhin
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Sudha K Shenoy
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA.
| |
Collapse
|
40
|
Wewer Albrechtsen NJ, Holst JJ, Cherrington AD, Finan B, Gluud LL, Dean ED, Campbell JE, Bloom SR, Tan TMM, Knop FK, Müller TD. 100 years of glucagon and 100 more. Diabetologia 2023; 66:1378-1394. [PMID: 37367959 DOI: 10.1007/s00125-023-05947-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/18/2023] [Indexed: 06/28/2023]
Abstract
The peptide hormone glucagon, discovered in late 1922, is secreted from pancreatic alpha cells and is an essential regulator of metabolic homeostasis. This review summarises experiences since the discovery of glucagon regarding basic and clinical aspects of this hormone and speculations on the future directions for glucagon biology and glucagon-based therapies. The review was based on the international glucagon conference, entitled 'A hundred years with glucagon and a hundred more', held in Copenhagen, Denmark, in November 2022. The scientific and therapeutic focus of glucagon biology has mainly been related to its role in diabetes. In type 1 diabetes, the glucose-raising properties of glucagon have been leveraged to therapeutically restore hypoglycaemia. The hyperglucagonaemia evident in type 2 diabetes has been proposed to contribute to hyperglycaemia, raising questions regarding underlying mechanism and the importance of this in the pathogenesis of diabetes. Mimicry experiments of glucagon signalling have fuelled the development of several pharmacological compounds including glucagon receptor (GCGR) antagonists, GCGR agonists and, more recently, dual and triple receptor agonists combining glucagon and incretin hormone receptor agonism. From these studies and from earlier observations in extreme cases of either glucagon deficiency or excess secretion, the physiological role of glucagon has expanded to also involve hepatic protein and lipid metabolism. The interplay between the pancreas and the liver, known as the liver-alpha cell axis, reflects the importance of glucagon for glucose, amino acid and lipid metabolism. In individuals with diabetes and fatty liver diseases, glucagon's hepatic actions may be partly impaired resulting in elevated levels of glucagonotropic amino acids, dyslipidaemia and hyperglucagonaemia, reflecting a new, so far largely unexplored pathophysiological phenomenon termed 'glucagon resistance'. Importantly, the hyperglucagonaemia as part of glucagon resistance may result in increased hepatic glucose production and hyperglycaemia. Emerging glucagon-based therapies show a beneficial impact on weight loss and fatty liver diseases and this has sparked a renewed interest in glucagon biology to enable further pharmacological pursuits.
Collapse
Affiliation(s)
- Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Lise Lotte Gluud
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - E Danielle Dean
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Endocrinology Division, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Stephen R Bloom
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M-M Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Filip K Knop
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| |
Collapse
|
41
|
Novikoff A, Müller TD. The molecular pharmacology of glucagon agonists in diabetes and obesity. Peptides 2023; 165:171003. [PMID: 36997003 PMCID: PMC10265134 DOI: 10.1016/j.peptides.2023.171003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Within recent decades glucagon receptor (GcgR) agonism has drawn attention as a therapeutic tool for the treatment of type 2 diabetes and obesity. In both mice and humans, glucagon administration enhances energy expenditure and suppresses food intake suggesting a promising metabolic utility. Therefore synthetic optimization of glucagon-based pharmacology to further resolve the physiological and cellular underpinnings mediating these effects has advanced. Chemical modifications to the glucagon sequence have allowed for greater peptide solubility, stability, circulating half-life, and understanding of the structure-function potential behind partial and "super"-agonists. The knowledge gained from such modifications has provided a basis for the development of long-acting glucagon analogues, chimeric unimolecular dual- and tri-agonists, and novel strategies for nuclear hormone targeting into glucagon receptor-expressing tissues. In this review, we summarize the developments leading toward the current advanced state of glucagon-based pharmacology, while highlighting the associated biological and therapeutic effects in the context of diabetes and obesity.
Collapse
Affiliation(s)
- Aaron Novikoff
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
42
|
Stenlid R, Manell H, Seth R, Cerenius SY, Chowdhury A, Roa Cortés C, Nyqvist I, Lundqvist T, Halldin M, Bergsten P. Low Fasting Concentrations of Glucagon in Patients with Very Long-Chain Acyl-CoA Dehydrogenase Deficiency. Metabolites 2023; 13:780. [PMID: 37512487 PMCID: PMC10386500 DOI: 10.3390/metabo13070780] [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: 05/23/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: Deficiencies of mitochondrial fatty acid oxidation (FAO) define a subgroup of inborn errors of metabolism, with medium-chain acyl-CoA dehydrogenase deficiency (MCAD) and very long-chain acyl-CoA dehydrogenase deficiency (VLCAD) being two of the most common. Hypoketotic hypoglycemia is a feared clinical complication and the treatment focuses on avoiding hypoglycemia. In contrast, carnitine uptake deficiency (CUD) is treated as a mild disease without significant effects on FAO. Impaired FAO has experimentally been shown to impair glucagon secretion. Glucagon is an important glucose-mobilizing hormone. If and how glucagon is affected in patients with VLCAD or MCAD remains unknown. (2) Methods: A cross-sectional study was performed with plasma hormone concentrations quantified after four hours of fasting. Patients with VLCAD (n = 10), MCAD (n = 7) and CUD (n = 6) were included. (3) Results: The groups were similar in age, sex, weight, and height. The glucagon and insulin levels were significantly lower in the VLCAD group compared to the CUD group (p < 0.05, respectively). The patients with CUD had glucagon concentrations similar to the normative data. No significant differences were seen in GLP-1, glicentin, glucose, amino acids, or NEFAs. (4) Conclusions: Low fasting concentrations of glucagon are present in patients with VLCAD and cannot be explained by altered stimuli in plasma.
Collapse
Affiliation(s)
- Rasmus Stenlid
- Department of Medical Cell Biology, Uppsala University, SE75123 Uppsala, Sweden
| | - Hannes Manell
- Department of Women's and Children's Health, Uppsala University, SE75185 Uppsala, Sweden
| | - Rikard Seth
- Department of Medical Cell Biology, Uppsala University, SE75123 Uppsala, Sweden
| | - Sara Y Cerenius
- Department of Medical Cell Biology, Uppsala University, SE75123 Uppsala, Sweden
| | - Azazul Chowdhury
- Department of Medical Cell Biology, Uppsala University, SE75123 Uppsala, Sweden
| | - Camilla Roa Cortés
- Department of Medical Cell Biology, Uppsala University, SE75123 Uppsala, Sweden
| | - Isabelle Nyqvist
- Department of Medical Cell Biology, Uppsala University, SE75123 Uppsala, Sweden
| | - Thomas Lundqvist
- Department of Women's and Children's Health, Karolinska Institute, SE17177 Stockholm, Sweden
| | - Maria Halldin
- Department of Women's and Children's Health, Karolinska Institute, SE17177 Stockholm, Sweden
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, SE75123 Uppsala, Sweden
| |
Collapse
|
43
|
Kaur S, Sokrat B, Capozzi ME, El K, Bai Y, Jazic A, Han B, Krishnakumar K, D'Alessio DA, Campbell JE, Bouvier M, Shenoy SK. The Ubiquitination Status of the Glucagon Receptor determines Signal Bias. J Biol Chem 2023; 299:104690. [PMID: 37037304 DOI: 10.1016/j.jbc.2023.104690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
The pancreatic hormone glucagon activates the glucagon receptor (GCGR), a class B seven-transmembrane G protein-coupled receptor (GPCR) that couples to the stimulatory heterotrimeric Gs protein and provokes protein kinase A-dependent signaling cascades vital to hepatic glucose metabolism and islet insulin secretion. Glucagon-stimulation also initiates recruitment of the endocytic adaptors, β-arrestin1 and β-arrestin2, which regulate desensitization and internalization of the GCGR. Unlike many other GPCRs, the GCGR expressed at the plasma membrane is constitutively ubiquitinated and upon agonist-activation, internalized GCGRs are deubiquitinated at early endosomes and recycled via Rab4-containing vesicles. Herein we report a novel link between the ubiquitination status and signal transduction mechanism of the GCGR. In the deubiquitinated state, coupling of the GCGR to Gs is diminished, while binding to β-arrestin is enhanced with signaling biased to a β-arrestin1-dependent p38 mitogen activated protein kinase (MAPK) pathway. This ubiquitin-dependent signaling bias arises through the modification of lysine333 (K333) on the cytoplasmic face of transmembrane helix V. Compared with the GCGR-WT, the mutant GCGR-K333R has impaired ubiquitination, diminished G protein coupling and protein kinase A signaling, but unimpaired potentiation of glucose-stimulated-insulin secretion in response to agonist-stimulation, which involves p38 MAPK signaling. Both WT and GCGR-K333R promote the formation of glucagon-induced β-arrestin1-dependent p38 signaling scaffold that requires canonical upstream MAPK-Kinase3, but is independent of Gs, Gi and β-arrestin2. Thus ubiquitination/deubiquitination at K333 in the GCGR defines the activation of distinct transducers with the potential to influence various facets of glucagon signaling in health and disease.
Collapse
Affiliation(s)
- Suneet Kaur
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Badr Sokrat
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, H3T 1J4 Canada; Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, H3T 1J4 Canada
| | - Megan E Capozzi
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Kimberley El
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Yushi Bai
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Aeva Jazic
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Bridgette Han
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Kaavya Krishnakumar
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford CA 94305
| | - David A D'Alessio
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Jonathan E Campbell
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, H3T 1J4 Canada; Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, H3T 1J4 Canada
| | - Sudha K Shenoy
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
44
|
Jungnik A, Arrubla Martinez J, Plum-Mörschel L, Kapitza C, Lamers D, Thamer C, Schölch C, Desch M, Hennige AM. Phase I studies of the safety, tolerability, pharmacokinetics and pharmacodynamics of the dual glucagon receptor/glucagon-like peptide-1 receptor agonist BI 456906. Diabetes Obes Metab 2023; 25:1011-1023. [PMID: 36527386 DOI: 10.1111/dom.14948] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/21/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
AIM To report two phase I studies of the novel subcutaneous glucagon-like peptide-1 receptor/glucagon receptor (GLP-1R/GCGR) dual agonist BI 456906 versus placebo in healthy volunteers and people with overweight/obesity. MATERIALS AND METHODS A phase Ia study (NCT03175211) investigated single rising doses (SRDs) of BI 456906 in 24 males with a body mass index (BMI) of 20-<30 kg/m2 . A phase Ib study (NCT03591718) investigated multiple rising doses (MRDs) of BI 456906 (escalated over 6 [Part A] or 16 [Part B] weeks) in 125 adults with a BMI of 27-40 kg/m2 . RESULTS In the SRD study (N = 24), mean body weight decreased with increasing BI 456906 dose. In the MRD study, the maximum decreases in placebo-corrected mean body weight were at week 6 (-5.79%, dosage schedule [DS] 1; Part A) and week 16 (-13.8%, DS7; Part B). BI 456906 reduced plasma amino acids and glucagon, indicating target engagement at GCGRs and GLP-1Rs. Drug-related adverse events (AEs) increased with BI 456906 dose. The most frequent drug-related AE with SRDs was decreased appetite (n = 9, 50.0%), and two subjects (8.3%) did not complete the trial because of AEs (nausea and vomiting). During MRD Part A (N = 80), 10 subjects (12.5%) discontinued BI 456906, most commonly because of a cardiac or vascular AE (n = 6, 7.5%); during Part B (N = 45), eight subjects (17.8%) discontinued BI 456906, mainly because of AEs (n = 6, 13.3%), most commonly gastrointestinal disorders. CONCLUSIONS BI 456906 produced a placebo-corrected body weight loss of 13.8% (week 16), highlighting its potential to promote clinically meaningful body weight loss in people with overweight/obesity.
Collapse
Affiliation(s)
| | | | | | - Christoph Kapitza
- Profil Institute for Metabolic Research, Neuss, Germany
- Profil Mainz GmbH & Co. KG, Mainz, Germany
| | | | - Claus Thamer
- Boehringer Ingelheim Pharma GmbH, Biberach, Germany
| | | | | | | |
Collapse
|
45
|
Doyle ME, Premathilake HU, Yao Q, Mazucanti CH, Egan JM. Physiology of the tongue with emphasis on taste transduction. Physiol Rev 2023; 103:1193-1246. [PMID: 36422992 PMCID: PMC9942923 DOI: 10.1152/physrev.00012.2022] [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] [Indexed: 11/25/2022] Open
Abstract
The tongue is a complex multifunctional organ that interacts and senses both interoceptively and exteroceptively. Although it is easily visible to almost all of us, it is relatively understudied and what is in the literature is often contradictory or is not comprehensively reported. The tongue is both a motor and a sensory organ: motor in that it is required for speech and mastication, and sensory in that it receives information to be relayed to the central nervous system pertaining to the safety and quality of the contents of the oral cavity. Additionally, the tongue and its taste apparatus form part of an innate immune surveillance system. For example, loss or alteration in taste perception can be an early indication of infection as became evident during the present global SARS-CoV-2 pandemic. Here, we particularly emphasize the latest updates in the mechanisms of taste perception, taste bud formation and adult taste bud renewal, and the presence and effects of hormones on taste perception, review the understudied lingual immune system with specific reference to SARS-CoV-2, discuss nascent work on tongue microbiome, as well as address the effect of systemic disease on tongue structure and function, especially in relation to taste.
Collapse
Affiliation(s)
- Máire E Doyle
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Hasitha U Premathilake
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Qin Yao
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Caio H Mazucanti
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Josephine M Egan
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
46
|
Hædersdal S, Andersen A, Knop FK, Vilsbøll T. Revisiting the role of glucagon in health, diabetes mellitus and other metabolic diseases. Nat Rev Endocrinol 2023; 19:321-335. [PMID: 36932176 DOI: 10.1038/s41574-023-00817-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 03/19/2023]
Abstract
Insulin and glucagon exert opposing effects on glucose metabolism and, consequently, pancreatic islet β-cells and α-cells are considered functional antagonists. The intra-islet hypothesis has previously dominated the understanding of glucagon secretion, stating that insulin acts to inhibit the release of glucagon. By contrast, glucagon is a potent stimulator of insulin secretion and has been used to test β-cell function. Over the past decade, α-cells have received increasing attention due to their ability to stimulate insulin secretion from neighbouring β-cells, and α-cell-β-cell crosstalk has proven central for glucose homeostasis in vivo. Glucagon is not only the counter-regulatory hormone to insulin in glucose metabolism but also glucagon secretion is more susceptible to changes in the plasma concentration of certain amino acids than to changes in plasma concentrations of glucose. Thus, the actions of glucagon also include a central role in amino acid turnover and hepatic fat oxidation. This Review provides insights into glucagon secretion, with a focus on the local paracrine actions on glucagon and the importance of α-cell-β-cell crosstalk. We focus on dysregulated glucagon secretion in obesity, non-alcoholic fatty liver disease and type 2 diabetes mellitus. Lastly, the future potential of targeting hyperglucagonaemia and applying dual and triple receptor agonists with glucagon receptor-activating properties in combination with incretin hormone receptor agonism is discussed.
Collapse
Affiliation(s)
- Sofie Hædersdal
- Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Herlev, Denmark.
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark.
| | - Andreas Andersen
- Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Herlev, Denmark
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Filip K Knop
- Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Herlev, Denmark
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Clinical Research, Copenhagen University Hospital - Steno Diabetes Center Copenhagen, Herlev, Denmark.
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark.
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
47
|
Kersten S. The impact of fasting on adipose tissue metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159262. [PMID: 36521736 DOI: 10.1016/j.bbalip.2022.159262] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/20/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Fasting and starvation were common occurrences during human evolution and accordingly have been an important environmental factor shaping human energy metabolism. Humans can tolerate fasting reasonably well through adaptative and well-orchestrated time-dependent changes in energy metabolism. Key features of the adaptive response to fasting are the breakdown of liver glycogen and muscle protein to produce glucose for the brain, as well as the gradual depletion of the fat stores, resulting in the release of glycerol and fatty acids into the bloodstream and the production of ketone bodies in the liver. In this paper, an overview is presented of our current understanding of the effects of fasting on adipose tissue metabolism. Fasting leads to reduced uptake of circulating triacylglycerols by adipocytes through inhibition of the activity of the rate-limiting enzyme lipoprotein lipase. In addition, fasting stimulates the degradation of stored triacylglycerols by activating the key enzyme adipose triglyceride lipase. The mechanisms underlying these events are discussed, with a special interest in insights gained from studies on humans. Furthermore, an overview is presented of the effects of fasting on other metabolic pathways in the adipose tissue, including fatty acid synthesis, glucose uptake, glyceroneogenesis, autophagy, and the endocrine function of adipose tissue.
Collapse
Affiliation(s)
- Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, the Netherlands.
| |
Collapse
|
48
|
Farag MR, Abo-Al-Ela HG, Alagawany M, Azzam MM, El-Saadony MT, Rea S, Di Cerbo A, Nouh DS. Effect of Quercetin Nanoparticles on Hepatic and Intestinal Enzymes and Stress-Related Genes in Nile Tilapia Fish Exposed to Silver Nanoparticles. Biomedicines 2023; 11:663. [PMID: 36979642 PMCID: PMC10045288 DOI: 10.3390/biomedicines11030663] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
Recently, nanotechnology has become an important research field involved in the improvement of animals' productivity, including aquaculture. In this field, silver nanoparticles (AgNPs) have gained interest as antibacterial, antiviral, and antifungal agents. On the other hand, their extensive use in other fields increased natural water pollution causing hazardous effects on aquatic organisms. Quercetin is a natural polyphenolic compound of many plants and vegetables, and it acts as a potent antioxidant and therapeutic agent in biological systems. The current study investigated the potential mitigative effect of quercetin nanoparticles (QNPs) against AgNPs-induced toxicity in Nile tilapia via investigating liver function markers, hepatic antioxidant status, apoptosis, and bioaccumulation of silver residues in hepatic tissue in addition to the whole-body chemical composition, hormonal assay, intestinal enzymes activity, and gut microbiota. Fish were grouped into: control fish, fish exposed to 1.98 mg L-1 AgNPs, fish that received 400 mg L-1 QNPs, and fish that received QNPs and AgNPs at the same concentrations. All groups were exposed for 60 days. The moisture and ash contents of the AgNP group were significantly higher than those of the other groups. In contrast, the crude lipid and protein decreased in the whole body. AgNPs significantly increased serum levels of ALT, AST, total cholesterol, and triglycerides and decreased glycogen and growth hormone (*** p < 0.001). The liver and intestinal enzymes' activities were significantly inhibited (*** p < 0.001), while the oxidative damage liver enzymes, intestinal bacterial and Aeromonas counts, and Ag residues in the liver were significantly increased (*** p < 0.001, and * p < 0.05). AgNPs also significantly upregulated the expression of hepatic Hsp70, caspase3, and p53 genes (* p < 0.05). These findings indicate the oxidative and hepatotoxic effects of AgNPs. QNPs enhanced and restored physiological parameters and health status under normal conditions and after exposure to AgNPs.
Collapse
Affiliation(s)
- Mayada R. Farag
- Forensic Medicine and Toxicology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt
| | - Haitham G. Abo-Al-Ela
- Genetics and Biotechnology, Department of Aquaculture, Faculty of Fish Resources, Suez University, Suez 43518, Egypt
| | - Mahmoud Alagawany
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig 44519, Egypt
| | - Mahmoud M. Azzam
- Department of Animal Production College of Food & Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed T. El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Stefano Rea
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
| | - Doaa S. Nouh
- Anatomy and Embryology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
49
|
Darbalaei S, Chang RL, Zhou QT, Chen Y, Dai AT, Wang MW, Yang DH. Effects of site-directed mutagenesis of GLP-1 and glucagon receptors on signal transduction activated by dual and triple agonists. Acta Pharmacol Sin 2023; 44:421-433. [PMID: 35953646 PMCID: PMC9889767 DOI: 10.1038/s41401-022-00962-y] [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: 04/21/2022] [Accepted: 07/17/2022] [Indexed: 02/04/2023] Open
Abstract
The paradigm of one drug against multiple targets, known as unimolecular polypharmacology, offers the potential to improve efficacy while overcoming some adverse events associated with the treatment. This approach is best exemplified by targeting two or three class B1 G protein-coupled receptors, namely, glucagon-like peptide-1 receptor (GLP-1R), glucagon receptor (GCGR) and glucose-dependent insulinotropic polypeptide receptor for treatment of type 2 diabetes and obesity. Some of the dual and triple agonists have already shown initial successes in clinical trials, although the molecular mechanisms underlying their multiplexed pharmacology remain elusive. In this study we employed structure-based site-directed mutagenesis together with pharmacological assays to compare agonist efficacy across two key signaling pathways, cAMP accumulation and ERK1/2 phosphorylation (pERK1/2). Three dual agonists (peptide 15, MEDI0382 and SAR425899) and one triple agonist (peptide 20) were evaluated at GLP-1R and GCGR, relative to the native peptidic ligands (GLP-1 and glucagon). Our results reveal the existence of residue networks crucial for unimolecular agonist-mediated receptor activation and their distinct signaling patterns, which might be useful to the rational design of biased drug leads.
Collapse
Affiliation(s)
- Sanaz Darbalaei
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ru-Lue Chang
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qing-Tong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yan Chen
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - An-Tao Dai
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
| | - Ming-Wei Wang
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Research Center for Deepsea Bioresources, Sanya, 572025, China.
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - De-Hua Yang
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Research Center for Deepsea Bioresources, Sanya, 572025, China.
| |
Collapse
|
50
|
Edgerton DS, Kraft G, Smith M, Farmer B, Williams P, Cherrington AD. A physiologic increase in brain glucagon action alters the hepatic gluconeogenic/glycogenolytic ratio but not glucagon's overall effect on glucose production. Am J Physiol Endocrinol Metab 2023; 324:E199-E208. [PMID: 36652399 PMCID: PMC9925168 DOI: 10.1152/ajpendo.00304.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
It has been proposed that brain glucagon action inhibits glucagon-stimulated hepatic glucose production (HGP), which may explain, at least in part, why glucagon's effect on HGP is transient. However, the pharmacologic off-target effects of glucagon in the brain may have been responsible for previously observed effects. Therefore, the aim of this study was to determine if central glucagon action plays a physiologic role in the regulation of HGP. Insulin was maintained at baseline while glucagon was either infused into the carotid and vertebral arteries or into a peripheral (leg) vein at rates designed to increase glucagon in the head in one group, while keeping glucagon at the liver matched between groups. The extraction rate of glucagon across the head was high (double that of the liver), and hypothalamic cAMP increased twofold, in proportion to the exposure of the brain to increased glucagon, but HGP was not reduced by the increase in brain glucagon signaling, as had been suggested previously (the areas under the curve for HGP were 840 ± 14 vs. 871 ± 36 mg/kg/240 min in head vs. peripheral infusion groups, respectively). Central nervous system glucagon action reduced circulating free fatty acids and glycerol, and this was associated with a modest reduction in net hepatic gluconeogenic flux. However, offsetting autoregulation by the liver (i.e., a reciprocal increase in net hepatic glycogenolysis) prevented a change in HGP. Thus, while physiologic engagement of the brain by glucagon can alter hepatic carbon flux, it does not appear to be responsible for the transient fall in HGP that occurs following the stimulation of HGP during a square wave rise in glucagon.NEW & NOTEWORTHY Glucagon stimulates hepatic glucose production through its direct effects on the liver but may indirectly inhibit this process by acting on the brain. This was tested by delivering glucagon via the cerebral circulatory system. Central nervous system glucagon action reduced liver gluconeogenic flux, but glycogenolysis increased, resulting in no net change in hepatic glucose production. Surprisingly, brain glucagon also appeared to suppress lipolysis (plasma free fatty acid and glycerol levels were reduced).
Collapse
Affiliation(s)
- Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Guillaume Kraft
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Marta Smith
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Ben Farmer
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Phillip Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| |
Collapse
|