1
|
Hong CT, Chen JH, Hu CJ. Role of glucagon-like peptide-1 receptor agonists in Alzheimer's disease and Parkinson's disease. J Biomed Sci 2024; 31:102. [PMID: 39501255 PMCID: PMC11539687 DOI: 10.1186/s12929-024-01090-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Neurodegenerative diseases, including Alzheimer's Disease (AD) and Parkinson's Disease (PD) are common complications of diabetes, arising from insulin resistance, inflammation, and other pathological processes in the central nervous system. The potential of numerous antidiabetic agents to modify neurodegenerative disease progression, both preclinically and clinically, has been assessed. These agents may provide additional therapeutic benefits beyond glycemic control. Introduced in the twenty-first century, glucagon-like peptide-1 receptor agonists (GLP-1RAs) are a class of antidiabetic drugs noted not only for their potent glucose-lowering effects but also for their cardiovascular and renal protective benefits. Various GLP-1RAs have been demonstrated to have significant benefits in in vitro and in vivo models of neurodegenerative diseases through modulating a variety of pathogenic mechanisms, including neuroinflammation, autophagy, mitochondrial dysfunction, and the abnormal phosphorylation of pathognomonic proteins. These agents also have substantial protective effects on cognitive and behavioral functions, such as motor function. However, clinical trials investigating GLP-1RAs in diseases such as AD, PD, mild cognitive impairment, psychiatric disorders, and diabetes have yielded mixed results for cognitive and motor function. This review examines the link between diabetes and neurodegenerative diseases, explores the effects of antidiabetic agents on neurodegeneration, provides a concise overview of the GLP-1 pathway, and discusses both preclinical and clinical trial outcomes of GLP-1RAs for neurodegenerative diseases, including their effects on cognition in AD and PD. This review also proposed new strategies for the design of future clinical trials on GLP-1 RAs for both AD and PD.
Collapse
Affiliation(s)
- Chien-Tai Hong
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wuxing St., Xinyi Dist., Taipei, 110, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Jia-Hung Chen
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wuxing St., Xinyi Dist., Taipei, 110, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Chaur-Jong Hu
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wuxing St., Xinyi Dist., Taipei, 110, Taiwan.
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Krishnan G, Bagath M, Devaraj C, Soren NM. The signalling association of glucagon-like peptide-1 and its receptors in the gastrointestinal tract and GPR40 and insulin receptor in the pancreas of sheep. Gen Comp Endocrinol 2024; 358:114602. [PMID: 39226991 DOI: 10.1016/j.ygcen.2024.114602] [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: 04/08/2024] [Revised: 08/19/2024] [Accepted: 08/31/2024] [Indexed: 09/05/2024]
Abstract
The present study was aimed at gaining insight into the signalling relationship between glucagon-like peptide-1 (GLP-1) and its receptor (GLP-1R) in the regulation of glucose metabolism. Further, to assess the role of G-protein-coupled receptor 40 (GPR40) and insulin receptor (INSR) in the pancreas of sheep that were supplemented with calcium salts of long-chain fatty acids (CSFAs). An experiment was carried out over a period of 60 days with eighteen sheep, and they were fed with a standard basal diet. The sheep were divided into three groups: CSFA0 (without CSFAs), while CSFA3 and CSFA5 were supplemented with 3 % and 5 % of CSFAs, respectively. Plasma concentrations of GLP-1, insulin, glucagon, and glucose were assessed every two weeks. At the end of the experiment, sheep were slaughtered, and samples of gastrointestinal tract (GIT) epithelial tissues and pancreas were collected to assess the relative expression of mRNA of GPR40, GLP-1R, and INSR. Postprandial GLP-1 and insulin were increased by 3.7-4.1 and 1.45-1.5 times, respectively, in the CSFAs-supplemented groups compared to CSFA0. Post-feeding, glucagon and glucose levels decreased in CSFA3 and CSFA5 compared to CSFA0. The results indicated that the supplementation of LCFAs increased the expression of GLP-1R in the GIT and pancreas, as well as the mRNA of GPR40 and INSR in the pancreas. Chemosensing of LCFAs by GPR40 in the pancreas triggers signalling transduction, and enhanced GLP-1 and GLP-1R resulted in moderately increased insulin secretion and reduced glucagon levels. These combined effects, along with the glucose-lowering effect of GLP-1, effectively lowered glucose levels in normoglycemic sheep.
Collapse
Affiliation(s)
- G Krishnan
- Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India.
| | - M Bagath
- Animal Nutrition Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| | - C Devaraj
- Bioenergetics and Environmental Sciences Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| | - N M Soren
- Animal Nutrition Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| |
Collapse
|
3
|
Cho H, Lim J. The emerging role of gut hormones. Mol Cells 2024; 47:100126. [PMID: 39426686 DOI: 10.1016/j.mocell.2024.100126] [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: 09/16/2024] [Revised: 10/13/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024] Open
Abstract
The gut is traditionally recognized as the central organ for the digestion and absorption of nutrients, however, it also functions as a significant endocrine organ, secreting a variety of hormones such as glucagon-like peptide 1, serotonin, somatostatin, and glucocorticoids. These gut hormones, produced by specialized intestinal epithelial cells, are crucial not only for digestive processes but also for the regulation of a wide range of physiological functions, including appetite, metabolism, and immune responses. While gut hormones can exert systemic effects, they also play a pivotal role in maintaining local homeostasis within the gut. This review discusses the role of the gut as an endocrine organ, emphasizing the stimuli, the newly discovered functions, and the clinical significance of gut-secreted hormones. Deciphering the emerging role of gut hormones will lead to a better understanding of gut homeostasis, innovative treatments for disorders in the gut, as well as systemic diseases.
Collapse
Affiliation(s)
- Hyeryeong Cho
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaechul Lim
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
4
|
Rees TA, Buttle BJ, Tasma Z, Yang SH, Harris PWR, Walker CS. Tirzepatide, GIP(1-42) and GIP(1-30) display unique signaling profiles at two common GIP receptor variants, E354 and Q354. Front Pharmacol 2024; 15:1463313. [PMID: 39464637 PMCID: PMC11502443 DOI: 10.3389/fphar.2024.1463313] [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: 07/11/2024] [Accepted: 10/02/2024] [Indexed: 10/29/2024] Open
Abstract
Type 2 diabetes (T2D) and obesity are prevalent metabolic disorders affecting millions of individuals worldwide. A new effective therapeutic drug called tirzepatide for the treatment of obesity and T2D is a dual agonist of the GIP receptor and GLP-1 receptor. Tirzepatide is clinically more effective than GLP-1 receptor agonists but the reasons why are not well understood. Tirzepatide reportedly stimulates the GIP receptor more potently than the GLP-1 receptor. However, tirzepatide signaling has not been thoroughly investigated at the E354 (wildtype) or Q354 (E354Q) GIP receptor variants. The E354Q variant is associated increased risk of T2D and lower body mass index. To better understand GIP receptor signaling we characterized the activity of endogenous agonists and tirzepatide at both GIP receptor variants. Using Cos7 cells we examined wildtype and E354Q GIP receptor signaling, analyzing cAMP and IP1 accumulation as well as AKT, ERK1/2 and CREB phosphorylation. GIP(1-42) and GIP(1-30)NH2 displayed equipotent effects on these pathways excluding CREB phosphorylation where GIP(1-30)NH2 was more potent than GIP(1-42) at the E354Q GIP receptor. Tirzepatide favored cAMP signaling at both variants. These findings indicate that tirzepatide is a biased agonist towards Gαs signaling and suggests it equally activates the wildtype and E354Q GIP receptor variants. We also observed differences between the pharmacology of the GIP receptor variants with endogenous peptides, which may help to explain differences in phenotype. These findings contribute to a comprehensive understanding of GIP receptor signaling, and will aid development of therapies combating T2D and obesity.
Collapse
Affiliation(s)
- Tayla A. Rees
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
- Headache Group, Wolfson Sensory Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Benjamin J. Buttle
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Zoe Tasma
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Sung-Hyun Yang
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Paul W. R. Harris
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Christopher S. Walker
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
5
|
Wang YX, Pi JC, Yao YF, Peng XP, Li WJ, Xie MY. Hypoglycemic effects of white hyacinth bean polysaccharide on type 2 diabetes mellitus rats involvement with entero-insular axis and GLP-1 via metabolomics study. Int J Biol Macromol 2024; 281:136489. [PMID: 39393741 DOI: 10.1016/j.ijbiomac.2024.136489] [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: 05/06/2024] [Revised: 09/02/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
The present study aimed to investigate the potential effects of white hyacinth bean polysaccharide (WHBP) against type 2 diabetic mellitus (T2DM) which was established by high-glucose/high-fat for 8 weeks, combined with a low-dose streptozotocin (STZ) injection. Our results showed that WHBP behaved the hypoglycemic effect by attenuating fasting blood glucose in vivo. WHBP-mediated anti-diabetic effects associated with the attenuation of insulin resistance and pancreatic impairment, as evidenced by the mitigation of pathological changes, inflammatory response and oxidative stress in the pancreas of T2DM rats. Meanwhile, gut protection was also shown during WHBP-mediated anti-diabetic effects, and glucagon-like peptide-1 (GLP-1), a mediator of the entero-insular axis, was observed to be elevated in both gut and pancreas of WHBP groups when compared to DM group, suggesting that hypoglycemic effects of WHBP were implicated in gut-pancreas interaction. Subsequently, untargeted metabolomics analysis performed by UPLC-QTOF/MS and showed that WHBP administration significantly adjusted the levels of 40 metabolites when compared to DM group. Further data concerning pathway analysis showed that WHBP administration significantly regulated the phenylalanine metabolism, tryptophan metabolism, arginine and proline, isoleucine metabolism, and glycerophospholipid metabolism in T2DM rats. Together, our results suggested that WHBP performed hypoglycemic effects and pancreatic protection linked to entero-insular axis involvement with GLP-1 and reversed metabolic disturbances in T2DM rats.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jin-Chan Pi
- College of Future Technology, Nanchang University, Nanchang 330031, China
| | - Yu-Fei Yao
- Department of Critical Care Medicine, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
| | - Xiao-Ping Peng
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Wen-Juan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| | - Ming-Yong Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
6
|
Okuma H, Tsuchiya K. Tissue-specific activation of insulin signaling as a potential target for obesity-related metabolic disorders. Pharmacol Ther 2024; 262:108699. [PMID: 39111411 DOI: 10.1016/j.pharmthera.2024.108699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
The incidence of obesity is rapidly increasing worldwide. Obesity-associated insulin resistance has long been established as a significant risk factor for obesity-related disorders such as type 2 diabetes and atherosclerosis. Insulin plays a key role in systemic glucose metabolism, with the liver, skeletal muscle, and adipose tissue as the major acting tissues. Insulin receptors and the downstream insulin signaling-related molecules are expressed in various tissues, including vascular endothelial cells, vascular smooth muscle cells, and monocytes/macrophages. In obesity, decreased insulin action is considered a driver for associated disorders. However, whether insulin action has a positive or negative effect on obesity-related disorders depends on the tissue in which it acts. While an enhancement of insulin signaling in the liver increases hepatic fat accumulation and exacerbates dyslipidemia, enhancement of insulin signaling in adipose tissue protects against obesity-related dysfunction of various organs by increasing the capacity for fat accumulation in the adipose tissue and inhibiting ectopic fat accumulation. Thus, this "healthy adipose tissue expansion" by enhancing insulin sensitivity in adipose tissue, but not in the liver, may be an effective therapeutic strategy for obesity-related disorders. To effectively address obesity-related metabolic disorders, the mechanisms of insulin resistance in various tissues of obese patients must be understood and drugs that enhance insulin action must be developed. In this article, we review the potential of interventions that enhance insulin signaling as a therapeutic strategy for obesity-related disorders, focusing on the molecular mechanisms of insulin action in each tissue.
Collapse
Affiliation(s)
- Hideyuki Okuma
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan
| | - Kyoichiro Tsuchiya
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan.
| |
Collapse
|
7
|
Huang HH, Wang YJ, Jiang HY, Yu HW, Chen YQ, Chiou A, Kuo JC. Sarcopenia-related changes in serum GLP-1 level affect myogenic differentiation. J Cachexia Sarcopenia Muscle 2024; 15:1708-1721. [PMID: 38926763 PMCID: PMC11446708 DOI: 10.1002/jcsm.13524] [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: 11/24/2023] [Revised: 04/26/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Sarcopenia, a group of muscle-related disorders, leads to the gradual decline and weakening of skeletal muscle over time. Recognizing the pivotal role of gastrointestinal conditions in maintaining metabolic homeostasis within skeletal muscle, we hypothesize that the effectiveness of the myogenic programme is influenced by the levels of gastrointestinal hormones in the bloodstream, and this connection is associated with the onset of sarcopenia. METHODS We first categorized 145 individuals from the Emergency Room of Taipei Veterans General Hospital into sarcopenia and non-sarcopenia groups, following the criteria established by the Asian Working Group for Sarcopenia. A thorough examination of specific gastrointestinal hormone levels in plasma was conducted to identify the one most closely associated with sarcopenia. Techniques, including immunofluorescence, western blotting, glucose uptake assays, seahorse real-time cell metabolic analysis, flow cytometry analysis, kinesin-1 activity assays and qPCR analysis, were applied to investigate its impacts and mechanisms on myogenic differentiation. RESULTS Individuals in the sarcopenia group exhibited elevated plasma levels of glucagon-like peptide 1 (GLP-1) at 1021.5 ± 313.5 pg/mL, in contrast to non-sarcopenic individuals with levels at 351.1 ± 39.0 pg/mL (P < 0.05). Although it is typical for GLP-1 levels to rise post-meal and subsequently drop naturally, detecting higher GLP-1 levels in starving individuals with sarcopenia raised the possibility of GLP-1 influencing myogenic differentiation in skeletal muscle. Further investigation using a cell model revealed that GLP-1 (1, 10 and 100 ng/mL) dose-dependently suppressed the expression of the myogenic marker, impeding myocyte fusion and the formation of polarized myotubes during differentiation. GLP-1 significantly inhibited the activity of the microtubule motor kinesin-1, interfering with the translocation of glucose transporter 4 (GLUT4) to the cell membrane and the dispersion of mitochondria. These impairments subsequently led to a reduction in glucose uptake to 0.81 ± 0.04 fold (P < 0.01) and mitochondrial adenosine triphosphate (ATP) production from 25.24 ± 1.57 pmol/min to 18.83 ± 1.11 pmol/min (P < 0.05). Continuous exposure to GLP-1, even under insulin induction, attenuated the elevated glucose uptake. CONCLUSIONS The elevated GLP-1 levels observed in individuals with sarcopenia are associated with a reduction in myogenic differentiation. The impact of GLP-1 on both the membrane translocation of GLUT4 and the dispersion of mitochondria significantly hinders glucose uptake and the production of mitochondrial ATP necessary for the myogenic programme. These findings point us towards strategies to establish the muscle-gut axis, particularly in the context of sarcopenia. Additionally, these results present the potential of identifying relevant diagnostic biomarkers.
Collapse
Affiliation(s)
- Hsien-Hao Huang
- Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yun-Jie Wang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hui-Yu Jiang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Helen Wenshin Yu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yin-Quan Chen
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Arthur Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
8
|
Zheng Z, Zong Y, Ma Y, Tian Y, Pang Y, Zhang C, Gao J. Glucagon-like peptide-1 receptor: mechanisms and advances in therapy. Signal Transduct Target Ther 2024; 9:234. [PMID: 39289339 PMCID: PMC11408715 DOI: 10.1038/s41392-024-01931-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/17/2024] [Accepted: 07/16/2024] [Indexed: 09/19/2024] Open
Abstract
The glucagon-like peptide-1 (GLP-1) receptor, known as GLP-1R, is a vital component of the G protein-coupled receptor (GPCR) family and is found primarily on the surfaces of various cell types within the human body. This receptor specifically interacts with GLP-1, a key hormone that plays an integral role in regulating blood glucose levels, lipid metabolism, and several other crucial biological functions. In recent years, GLP-1 medications have become a focal point in the medical community due to their innovative treatment mechanisms, significant therapeutic efficacy, and broad development prospects. This article thoroughly traces the developmental milestones of GLP-1 drugs, from their initial discovery to their clinical application, detailing the evolution of diverse GLP-1 medications along with their distinct pharmacological properties. Additionally, this paper explores the potential applications of GLP-1 receptor agonists (GLP-1RAs) in fields such as neuroprotection, anti-infection measures, the reduction of various types of inflammation, and the enhancement of cardiovascular function. It provides an in-depth assessment of the effectiveness of GLP-1RAs across multiple body systems-including the nervous, cardiovascular, musculoskeletal, and digestive systems. This includes integrating the latest clinical trial data and delving into potential signaling pathways and pharmacological mechanisms. The primary goal of this article is to emphasize the extensive benefits of using GLP-1RAs in treating a broad spectrum of diseases, such as obesity, cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), neurodegenerative diseases, musculoskeletal inflammation, and various forms of cancer. The ongoing development of new indications for GLP-1 drugs offers promising prospects for further expanding therapeutic interventions, showcasing their significant potential in the medical field.
Collapse
Affiliation(s)
- Zhikai Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Yiyang Ma
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yucheng Tian
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
9
|
Perez-Frances M, Bru-Tari E, Cohrs C, Abate MV, van Gurp L, Furuyama K, Speier S, Thorel F, Herrera PL. Regulated and adaptive in vivo insulin secretion from islets only containing β-cells. Nat Metab 2024; 6:1791-1806. [PMID: 39169271 PMCID: PMC11422169 DOI: 10.1038/s42255-024-01114-8] [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: 04/26/2023] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
Insulin-producing β-cells in pancreatic islets are regulated by systemic cues and, locally, by adjacent islet hormone-producing 'non-β-cells' (namely α-cells, δ-cells and γ-cells). Yet whether the non-β-cells are required for accurate insulin secretion is unclear. Here, we studied mice in which adult islets are exclusively composed of β-cells and human pseudoislets containing only primary β-cells. Mice lacking non-β-cells had optimal blood glucose regulation, enhanced glucose tolerance, insulin sensitivity and restricted body weight gain under a high-fat diet. The insulin secretion dynamics in islets composed of only β-cells was comparable to that in intact islets. Similarly, human β-cell pseudoislets retained the glucose-regulated mitochondrial respiration, insulin secretion and exendin-4 responses of entire islets. The findings indicate that non-β-cells are dispensable for blood glucose homeostasis and β-cell function. These results support efforts aimed at developing diabetes treatments by generating β-like clusters devoid of non-β-cells, such as from pluripotent stem cells differentiated in vitro or by reprograming non-β-cells into insulin producers in situ.
Collapse
Affiliation(s)
- Marta Perez-Frances
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Eva Bru-Tari
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christian Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
| | - Maria Valentina Abate
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Léon van Gurp
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kenichiro Furuyama
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabrizio Thorel
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pedro L Herrera
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
10
|
Gonzalez E, Lee MD, Tierney BT, Lipieta N, Flores P, Mishra M, Beckett L, Finkelstein A, Mo A, Walton P, Karouia F, Barker R, Jansen RJ, Green SJ, Weging S, Kelliher J, Singh NK, Bezdan D, Galazska J, Brereton NJB. Spaceflight alters host-gut microbiota interactions. NPJ Biofilms Microbiomes 2024; 10:71. [PMID: 39209868 PMCID: PMC11362537 DOI: 10.1038/s41522-024-00545-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The ISS rodent habitat has provided crucial insights into the impact of spaceflight on mammals, inducing symptoms characteristic of liver disease, insulin resistance, osteopenia, and myopathy. Although these physiological responses can involve the microbiome on Earth, host-microbiota interactions during spaceflight are still being elucidated. We explore murine gut microbiota and host gene expression in the colon and liver after 29 and 56 days of spaceflight using multiomics. Metagenomics revealed significant changes in 44 microbiome species, including relative reductions in bile acid and butyrate metabolising bacteria like Extibacter muris and Dysosmobacter welbionis. Functional prediction indicate over-representation of fatty acid and bile acid metabolism, extracellular matrix interactions, and antibiotic resistance genes. Host gene expression described corresponding changes to bile acid and energy metabolism, and immune suppression. These changes imply that interactions at the host-gut microbiome interface contribute to spaceflight pathology and that these interactions might critically influence human health and long-duration spaceflight feasibility.
Collapse
Affiliation(s)
- E Gonzalez
- Microbiome Unit, Canadian Centre for Computational Genomics, Department of Human Genetics, McGill University, Montréal, Canada
- Centre for Microbiome Research, McGill University, Montréal, Canada
| | - M D Lee
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - B T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - N Lipieta
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - P Flores
- BioServe Space Technologies, University of Colorado Boulder, Boulder, CO, USA
| | - M Mishra
- Grossman School of Medicine, New York University, New York, USA
| | - L Beckett
- University of Nottingham, Nottingham, NG7 2RD, UK
| | - A Finkelstein
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - A Mo
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - P Walton
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - F Karouia
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Centre for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - R Barker
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Yuri GmbH, Wiesentalstr. 40, 88074, Meckenbeuren, Germany
- University of Wisconsin-Madison, Madison, WI, USA
| | - R J Jansen
- Department of Public Health, North Dakota State University, Fargo, ND, USA
- Genomics, Phenomics, and Bioinformatics Program, North Dakota State University, Fargo, ND, USA
| | - S J Green
- Genomics and Microbiome Core Facility, Rush University Medical Centre, 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - S Weging
- Institute of Computer Science, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - J Kelliher
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - N K Singh
- Department of Industrial Relations, Division of Occupational Safety and Health, Oakland, USA
| | - D Bezdan
- University of Wisconsin-Madison, Madison, WI, USA
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - J Galazska
- Space Biosciences Research Branch, NASA Ames Research Centre, Moffett Field, CA, USA
| | - N J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
11
|
Lian K, Zhang K, Kan C, Hou N, Han F, Sun X, Qiu H, Guo Z. Emerging therapeutic landscape: Incretin agonists in chronic kidney disease management. Life Sci 2024; 351:122801. [PMID: 38862060 DOI: 10.1016/j.lfs.2024.122801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024]
Abstract
The increasing incidence of chronic kidney disease (CKD) poses a significant public health concern, prompting heightened attention to its treatment. Incretins, including glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide, are intestinal peptides released after nutrient intake, known for their hypoglycemic effects in diabetes management. Recent advancements highlight the promising outcomes of GLP-1 receptor agonists in reducing CKD risk factors and improving renal outcomes. The multifaceted functions of GLP-1, such as its anti-obesity, anti-hypertensive, anti-hyperglycemic, anti-lipid, anti-inflammatory, and endothelial function protective properties, contribute to its potential as a therapeutic agent for CKD. Although experiments suggest the potential benefits of incretin in CKD, a comprehensive understanding of its specific mechanisms is still lacking. This review aims to provide a detailed examination of current evidence and potential future directions, emphasizing the promising yet evolving landscape of incretin agonists in the context of CKD.
Collapse
Affiliation(s)
- Kexin Lian
- Department of Nephropathy, Affiliated Hospital of Shandong Second Medical University, Weifang, China; Department of Endocrinology and Metabolism, Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China.
| | - Zhentao Guo
- Department of Nephropathy, Affiliated Hospital of Shandong Second Medical University, Weifang, China.
| |
Collapse
|
12
|
Zhao N, Wu G, Zhao L. H 2S as a metabolic saboteur. Nat Metab 2024; 6:1431-1432. [PMID: 39030388 DOI: 10.1038/s42255-024-01086-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Affiliation(s)
- Naisi Zhao
- Department of Public Health and Community Medicine, School of Medicine, Tufts University, Boston, MA, USA
| | - Guojun Wu
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences and Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Liping Zhao
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences and Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
- State Key Laboratory of Microbial Metabolism and Ministry of Education Key Laboratory of Systems Biomedicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
13
|
Bensignor MO, Arslanian S, Vajravelu ME. Semaglutide for management of obesity in adolescents: efficacy, safety, and considerations for clinical practice. Curr Opin Pediatr 2024; 36:449-455. [PMID: 38774967 PMCID: PMC11222026 DOI: 10.1097/mop.0000000000001365] [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] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review is to describe the existing limited data related to the use of semaglutide in adolescents with obesity, supplementing with findings from adult studies of semaglutide use. RECENT FINDINGS Semaglutide, as a once weekly subcutaneous injection for weight management, effectively reduces body mass index (BMI) while improving hyperglycemia, elevated alanine aminotransferase levels, hyperlipidemia, and quality of life in youth with obesity. As of this review, only one large randomized clinical trial of semaglutide in youth has been completed, with a follow-up duration of 68 weeks. Thus, long-term data on the safety in adolescents is limited, particularly regarding the risks of cholelithiasis, pancreatitis, suicidal ideation, and disordered eating. Due to the cost of semaglutide, particularly in the United States, limited cost effectiveness analyses have demonstrated unfavorable incremental cost-effectiveness ratios for semaglutide relative to phentermine-topiramate as an alternative antiobesity medication in adolescents. SUMMARY Semaglutide represents an important advance in the pediatric obesity management, with clear short-term reductions in BMI and improvement in metabolic parameters. However, its long-term safety and efficacy for youth with obesity remain to be demonstrated. Additional research is needed to assess trends in utilization and adherence to minimize the risk of worsening socioeconomic disparities in pediatric obesity.
Collapse
Affiliation(s)
- Megan O Bensignor
- Division of Pediatric Endocrinology and Diabetes, Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, Minnesota; Division of Pediatric Endocrinology and Diabetes, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Silva Arslanian
- Division of Pediatric Endocrinology, Diabetes, and Metabolism and Center for Pediatric Research in Obesity and Metabolism, UPMC Children's Hospital of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mary Ellen Vajravelu
- Division of Pediatric Endocrinology, Diabetes, and Metabolism and Center for Pediatric Research in Obesity and Metabolism, UPMC Children's Hospital of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Carrageta DF, Pereira SC, Ferreira R, Monteiro MP, Oliveira PF, Alves MG. Signatures of metabolic diseases on spermatogenesis and testicular metabolism. Nat Rev Urol 2024; 21:477-494. [PMID: 38528255 DOI: 10.1038/s41585-024-00866-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 03/27/2024]
Abstract
Diets leading to caloric overload are linked to metabolic disorders and reproductive function impairment. Metabolic and hormonal abnormalities stand out as defining features of metabolic disorders, and substantially affect the functionality of the testis. Metabolic disorders induce testicular metabolic dysfunction, chronic inflammation and oxidative stress. The disruption of gastrointestinal, pancreatic, adipose tissue and testicular hormonal regulation induced by metabolic disorders can also contribute to a state of compromised fertility. In this Review, we will delve into the effects of high-fat diets and metabolic disorders on testicular metabolism and spermatogenesis, which are crucial elements for male reproductive function. Moreover, metabolic disorders have been shown to influence the epigenome of male gametes and might have a potential role in transmitting phenotype traits across generations. However, the existing evidence strongly underscores the unmet need to understand the mechanisms responsible for transgenerational paternal inheritance of male reproductive function impairment related to metabolic disorders. This knowledge could be useful for developing targeted interventions to prevent, counteract, and most of all break the perpetuation chain of male reproductive dysfunction associated with metabolic disorders across generations.
Collapse
Affiliation(s)
- David F Carrageta
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Sara C Pereira
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Mariana P Monteiro
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Marco G Alves
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Campus de Santiago Agra do Crasto, Aveiro, Portugal.
| |
Collapse
|
15
|
Abiola JO, Oluyemi AA, Idowu OT, Oyinloye OM, Ubah CS, Owolabi OV, Somade OT, Onikanni SA, Ajiboye BO, Osunsanmi FO, Nash O, Omotuyi OI, Oyinloye BE. Potential Role of Phytochemicals as Glucagon-like Peptide 1 Receptor (GLP-1R) Agonists in the Treatment of Diabetes Mellitus. Pharmaceuticals (Basel) 2024; 17:736. [PMID: 38931402 PMCID: PMC11206448 DOI: 10.3390/ph17060736] [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: 04/07/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Currently, there is no known cure for diabetes. Different pharmaceutical therapies have been approved for the management of type 2 diabetes mellitus (T2DM), some are in clinical trials and they have been classified according to their route or mechanism of action. Insulin types, sulfonylureas, biguanides, alpha-glucosidase inhibitors, thiazolidinediones, meglitinides, sodium-glucose cotransporter type 2 inhibitors, and incretin-dependent therapies (glucagon-like peptide-1 receptor agonists: GLP-1R, and dipeptidyl peptidase 4 inhibitors: DPP-4). Although some of the currently available drugs are effective in the management of T2DM, the side effects resulting from prolonged use of these drugs remain a serious challenge. GLP-1R agonists are currently the preferred medications to include when oral metformin alone is insufficient to manage T2DM. Medicinal plants now play prominent roles in the management of various diseases globally because they are readily available and affordable as well as having limited and transient side effects. Recently, studies have reported the ability of phytochemicals to activate glucagon-like peptide-1 receptor (GLP-1R), acting as an agonist just like the GLP-1R agonist with beneficial effects in the management of T2DM. Consequently, we propose that careful exploration of phytochemicals for the development of novel therapeutic candidates as GLP-1R agonists will be a welcome breakthrough in the management of T2DM and the co-morbidities associated with T2DM.
Collapse
Affiliation(s)
- Julianah Ore Abiola
- Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, Ado-Ekiti 360001, Nigeria; (J.O.A.)
- Center for Genomics Research and Innovation, National Biotechnology Development Agency, Abuja 09004, Nigeria
- Institute of Drug Research and Development, S.E. Bogoro Center, Afe Babalola University, Ado-Ekiti 360001, Nigeria
| | - Ayoola Abidemi Oluyemi
- Institute of Drug Research and Development, S.E. Bogoro Center, Afe Babalola University, Ado-Ekiti 360001, Nigeria
| | - Olajumoke Tolulope Idowu
- Industrial Chemistry Unit, Department of Chemical Sciences, College of Sciences, Afe Babalola University, Ado-Ekiti 360001, Nigeria
| | - Oluwatoyin Mary Oyinloye
- Department of Mathematics, Science and Technology Education, Faculty of Education, University of Zululand, Kwadlangezwa 3886, South Africa
| | - Chukwudi Sunday Ubah
- Department of Epidemiology and Biostatistics, College of Public Health, Temple University, Philadelphia, PA 19121, USA
| | - Olutunmise Victoria Owolabi
- Medical Biochemistry Unit, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti 360001, Nigeria
| | - Oluwatobi T. Somade
- Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, Ado-Ekiti 360001, Nigeria; (J.O.A.)
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta 111101, Nigeria
| | - Sunday Amos Onikanni
- Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, Ado-Ekiti 360001, Nigeria; (J.O.A.)
- College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Basiru Olaitan Ajiboye
- Institute of Drug Research and Development, S.E. Bogoro Center, Afe Babalola University, Ado-Ekiti 360001, Nigeria
- Phytomedicine and Molecular Toxicology Research Laboratory, Department of Biochemistry, Federal University Oye-Ekiti, Oye-Ekiti 371104, Nigeria
| | - Foluso Oluwagbemiga Osunsanmi
- Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, Kwadlangezwa 3886, South Africa
| | - Oyekanmi Nash
- Center for Genomics Research and Innovation, National Biotechnology Development Agency, Abuja 09004, Nigeria
| | - Olaposi Idowu Omotuyi
- Institute of Drug Research and Development, S.E. Bogoro Center, Afe Babalola University, Ado-Ekiti 360001, Nigeria
- Department of Pharmacology and Toxicology, College of Pharmacy, Afe Babalola University, Ado-Ekiti 360001, Nigeria
| | - Babatunji Emmanuel Oyinloye
- Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, Ado-Ekiti 360001, Nigeria; (J.O.A.)
- Institute of Drug Research and Development, S.E. Bogoro Center, Afe Babalola University, Ado-Ekiti 360001, Nigeria
- Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, Kwadlangezwa 3886, South Africa
| |
Collapse
|
16
|
Yang Z, Chen S, Sun W, Yang Y, Xu Y, Tang Y, Jiang W, Li J, Zhang Y. Study on the mechanisms by which pumpkin polysaccharides regulate abnormal glucose and lipid metabolism in diabetic mice under oxidative stress. Int J Biol Macromol 2024; 270:132249. [PMID: 38729500 DOI: 10.1016/j.ijbiomac.2024.132249] [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: 11/04/2023] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Pumpkin polysaccharide (PPe-H) can perform physiological functions through its antioxidative and hypoglycemic effects; however, the mechanisms through which PPe-H regulates abnormal glucose and lipid metabolism caused by oxidative stress injury remain unclear. In the present study, streptozotocin was used to generate an acute diabetic mouse model, and the effects of PPe-H on glucose and lipid metabolism impaired by oxidative stress in diabetic mice were studied. PPe-H significantly reduced blood glucose levels and enhanced the oral glucose tolerance of diabetic mice under stress injury (p < 0.05). The analysis of liver antioxidant enzymes showed that PPe-H significantly enhanced the activities of SOD and CAT (p < 0.05), increased the GSH level, and decreased the level of MDA (p < 0.05). Transcriptomic and metabolomic analyses of the liver tissues of mice revealed characteristic differences in the genetic and metabolic levels of the samples, which showed that PPe-H treatment may play a positive role in regulating the metabolism of methionine, cysteine, glycerol phospholipid, and linoleic acid. These results indicated that PPe-H alleviated the symptoms of hyperglycemia by regulating metabolites related to oxidative stress and glycolipid metabolism in diabetic mice.
Collapse
Affiliation(s)
- Zeen Yang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Shengdong Chen
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Wenxuan Sun
- Liang Xin College, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Yechen Yang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Yuxuan Xu
- Liang Xin College, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Yuxuan Tang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Wen Jiang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Jia Li
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China
| | - Yongjun Zhang
- College of Life Sciences, Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, China Jiliang University, Hangzhou, Zhejiang Province 310018, China.
| |
Collapse
|
17
|
Azmy Nabeh O, Amr A, Faoosa AM, Esmat E, Osama A, Khedr AS, Amin B, Saud AI, Elmorsy SA. Emerging Perspectives on the Impact of Diabetes Mellitus and Anti-Diabetic Drugs on Premenstrual Syndrome. A Narrative Review. Diabetes Ther 2024; 15:1279-1299. [PMID: 38668996 PMCID: PMC11096298 DOI: 10.1007/s13300-024-01585-8] [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: 03/15/2024] [Accepted: 04/08/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetes mellitus (DM) and premenstrual syndrome (PMS) are global health challenges. Both disorders are often linked to a range of physical and psychological symptoms that significantly impact the quality of life of many women. Yet, the exact relation between DM and PMS is not clear, and the management of both conditions poses a considerable challenge. In this review, we aimed to investigate the interplay between DM, anti-diabetic drugs, and the different theories and symptoms of PMS. Female sex hormones are implicated in the pathophysiology of PMS and can also impair blood glucose control. In addition, patients with diabetes face a higher susceptibility to anxiety and depression disorders, with a significant number of patients experiencing symptoms such as fatigue and difficulty concentrating, which are reported in patients with PMS as well. Complications related to diabetic medications, such as hypoglycemia (with sulfonylurea) and fluid retention (with thiazolidinediones) may also mediate PMS-like symptoms. DM can, in addition, disturb the normal gut microbiota (GM), with a consequent loss of beneficial GM metabolites that guard against PMS, particularly the short-chain fatty acids and serotonin. Among the several available anti-diabetic drugs, those (1) with an anti-inflammatory potential, (2) that can preserve the beneficial GM, and (3) possessing a lower risk for hypoglycemia, might have a favorable outcome in PMS women. Yet, well-designed clinical trials are needed to investigate the anti-diabetic drug(s) of choice for patients with diabetes and PMS.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Alaa Amr
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Eshraka Esmat
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa Osama
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Basma Amin
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa I Saud
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
18
|
Zhang C, Shi Y, Liu C, Sudesh SM, Hu Z, Li P, Liu Q, Ma Y, Shi A, Cai H. Therapeutic strategies targeting mechanisms of macrophages in diabetic heart disease. Cardiovasc Diabetol 2024; 23:169. [PMID: 38750502 PMCID: PMC11097480 DOI: 10.1186/s12933-024-02273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 05/08/2024] [Indexed: 05/18/2024] Open
Abstract
Diabetic heart disease (DHD) is a serious complication in patients with diabetes. Despite numerous studies on the pathogenic mechanisms and therapeutic targets of DHD, effective means of prevention and treatment are still lacking. The pathogenic mechanisms of DHD include cardiac inflammation, insulin resistance, myocardial fibrosis, and oxidative stress. Macrophages, the primary cells of the human innate immune system, contribute significantly to these pathological processes, playing an important role in human disease and health. Therefore, drugs targeting macrophages hold great promise for the treatment of DHD. In this review, we examine how macrophages contribute to the development of DHD and which drugs could potentially be used to target macrophages in the treatment of DHD.
Collapse
Affiliation(s)
- Chaoyue Zhang
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunke Shi
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Changzhi Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shivon Mirza Sudesh
- Faculty of Medicine, St. George University of London, London, UK
- University of Nicosia Medical School, University of Nicosia, Nicosia, Cyprus
| | - Zhao Hu
- Department of Geriatric Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pengyang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Qi Liu
- Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Yiming Ma
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ao Shi
- Faculty of Medicine, St. George University of London, London, UK.
- University of Nicosia Medical School, University of Nicosia, Nicosia, Cyprus.
| | - Hongyan Cai
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
19
|
Zhang L, Wang P, Huang J, Xing Y, Wong FS, Suo J, Wen L. Gut microbiota and therapy for obesity and type 2 diabetes. Front Endocrinol (Lausanne) 2024; 15:1333778. [PMID: 38596222 PMCID: PMC11002083 DOI: 10.3389/fendo.2024.1333778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/06/2024] [Indexed: 04/11/2024] Open
Abstract
There has been a major increase in Type 2 diabetes and obesity in many countries, and this will lead to a global public health crisis, which not only impacts on the quality of life of individuals well but also places a substantial burden on healthcare systems and economies. Obesity is linked to not only to type 2 diabetes but also cardiovascular diseases, musculoskeletal disorders, and certain cancers, also resulting in increased medical costs and diminished quality of life. A number of studies have linked changes in gut in obesity development. Dysbiosis, a deleterious change in gut microbiota composition, leads to altered intestinal permeability, associated with obesity and Type 2 diabetes. Many factors affect the homeostasis of gut microbiota, including diet, genetics, circadian rhythms, medication, probiotics, and antibiotics. In addition, bariatric surgery induces changes in gut microbiota that contributes to the metabolic benefits observed post-surgery. Current obesity management strategies encompass dietary interventions, exercise, pharmacotherapy, and bariatric surgery, with emerging treatments including microbiota-altering approaches showing promising efficacy. While pharmacotherapy has demonstrated significant advancements in recent years, bariatric surgery remains one of the most effective treatments for sustainable weight loss. However, access to this is generally limited to those living with severe obesity. This underscores the need for non-surgical interventions, particularly for adolescents and mildly obese patients. In this comprehensive review, we assess longitudinal alterations in gut microbiota composition and functionality resulting from the two currently most effective anti-obesity treatments: pharmacotherapy and bariatric surgery. Additionally, we highlight the functions of gut microbiota, focusing on specific bacteria, their metabolites, and strategies for modulating gut microbiota to prevent and treat obesity. This review aims to provide insights into the evolving landscape of obesity management and the potential of microbiota-based approaches in addressing this pressing global health challenge.
Collapse
Affiliation(s)
- Luyao Zhang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Pai Wang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Juan Huang
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha, Hunan, China
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanpeng Xing
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - F. Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jian Suo
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
20
|
Varney MJ, Benovic JL. The Role of G Protein-Coupled Receptors and Receptor Kinases in Pancreatic β-Cell Function and Diabetes. Pharmacol Rev 2024; 76:267-299. [PMID: 38351071 PMCID: PMC10877731 DOI: 10.1124/pharmrev.123.001015] [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: 08/03/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 02/16/2024] Open
Abstract
Type 2 diabetes (T2D) mellitus has emerged as a major global health concern that has accelerated in recent years due to poor diet and lifestyle. Afflicted individuals have high blood glucose levels that stem from the inability of the pancreas to make enough insulin to meet demand. Although medication can help to maintain normal blood glucose levels in individuals with chronic disease, many of these medicines are outdated, have severe side effects, and often become less efficacious over time, necessitating the need for insulin therapy. G protein-coupled receptors (GPCRs) regulate many physiologic processes, including blood glucose levels. In pancreatic β cells, GPCRs regulate β-cell growth, apoptosis, and insulin secretion, which are all critical in maintaining sufficient β-cell mass and insulin output to ensure euglycemia. In recent years, new insights into the signaling of incretin receptors and other GPCRs have underscored the potential of these receptors as desirable targets in the treatment of diabetes. The signaling of these receptors is modulated by GPCR kinases (GRKs) that phosphorylate agonist-activated GPCRs, marking the receptor for arrestin binding and internalization. Interestingly, genome-wide association studies using diabetic patient cohorts link the GRKs and arrestins with T2D. Moreover, recent reports show that GRKs and arrestins expressed in the β cell serve a critical role in the regulation of β-cell function, including β-cell growth and insulin secretion in both GPCR-dependent and -independent pathways. In this review, we describe recent insights into GPCR signaling and the importance of GRK function in modulating β-cell physiology. SIGNIFICANCE STATEMENT: Pancreatic β cells contain a diverse array of G protein-coupled receptors (GPCRs) that have been shown to improve β-cell function and survival, yet only a handful have been successfully targeted in the treatment of diabetes. This review discusses recent advances in our understanding of β-cell GPCR pharmacology and regulation by GPCR kinases while also highlighting the necessity of investigating islet-enriched GPCRs that have largely been unexplored to unveil novel treatment strategies.
Collapse
Affiliation(s)
- Matthew J Varney
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Lai TT, Tsai YH, Liou CW, Fan CH, Hou YT, Yao TH, Chuang HL, Wu WL. The gut microbiota modulate locomotion via vagus-dependent glucagon-like peptide-1 signaling. NPJ Biofilms Microbiomes 2024; 10:2. [PMID: 38228675 DOI: 10.1038/s41522-024-00477-w] [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: 04/25/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024] Open
Abstract
Locomotor activity is an innate behavior that can be triggered by gut-motivated conditions, such as appetite and metabolic condition. Various nutrient-sensing receptors distributed in the vagal terminal in the gut are crucial for signal transduction from the gut to the brain. The levels of gut hormones are closely associated with the colonization status of the gut microbiota, suggesting a complicated interaction among gut bacteria, gut hormones, and the brain. However, the detailed mechanism underlying gut microbiota-mediated endocrine signaling in the modulation of locomotion is still unclear. Herein, we show that broad-spectrum antibiotic cocktail (ABX)-treated mice displayed hypolocomotion and elevated levels of the gut hormone glucagon-like peptide-1 (GLP-1). Blockade of the GLP-1 receptor and subdiaphragmatic vagal transmission rescued the deficient locomotor phenotype in ABX-treated mice. Activation of the GLP-1 receptor and vagal projecting brain regions led to hypolocomotion. Finally, selective antibiotic treatment dramatically increased serum GLP-1 levels and decreased locomotion. Colonizing Lactobacillus reuteri and Bacteroides thetaiotaomicron in microbiota-deficient mice suppressed GLP-1 levels and restored the hypolocomotor phenotype. Our findings identify a mechanism by which specific gut microbes mediate host motor behavior via the enteroendocrine and vagal-dependent neural pathways.
Collapse
Affiliation(s)
- Tzu-Ting Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Hsuan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Chia-Wei Liou
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Tian Hou
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Tzu-Hsuan Yao
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, 115202, Taiwan
| | - Wei-Li Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|
22
|
Groenen C, Nguyen TA, Paulusma C, van de Graaf S. Bile salt signaling and bile salt-based therapies in cardiometabolic disease. Clin Sci (Lond) 2024; 138:1-21. [PMID: 38180064 PMCID: PMC10767275 DOI: 10.1042/cs20230934] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 01/06/2024]
Abstract
Bile salts have an established role in the emulsification and intestinal absorption of dietary lipids, and their homeostasis is tightly controlled by various transporters and regulators in the enterohepatic circulation. Notably, emerging evidence points toward bile salts as major modulators of cardiometabolic disease (CMD), an umbrella disease of disorders affecting the heart and blood vessels that is caused by systemic metabolic diseases such as Type 2 diabetes mellitus (T2DM) and metabolic dysfunction-associated steatotic liver disease (MASLD), the latter encompassing also metabolic dysfunction-associated steatohepatitis (MASH). The underlying mechanisms of protective effects of bile salts are their hormonal properties, enabling them to exert versatile metabolic effects by activating various bile salt-responsive signaling receptors with the nuclear farnesoid X receptor (FXR) and the Takeda G-protein-coupled receptor 5 (TGR5) as most extensively investigated. Activation of FXR and TGR5 is involved in the regulation of glucose, lipid and energy metabolism, and inflammation. Bile salt-based therapies directly targeting FXR and TGR5 signaling have been evaluated for their therapeutic potential in CMD. More recently, therapeutics targeting bile salt transporters thereby modulating bile salt localization, dynamics, and signaling, have been developed and evaluated in CMD. Here, we discuss the current knowledge on the contribution of bile salt signaling in the pathogenesis of CMD and the potential of bile salt-based therapies for the treatment of CMD.
Collapse
Affiliation(s)
- Claire C.J. Groenen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, The Netherlands
| | - Thuc-Anh Nguyen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, The Netherlands
| | - Coen C. Paulusma
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, The Netherlands
| | - Stan F.J. van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, The Netherlands
| |
Collapse
|
23
|
Bisht S, Singh MF. The triggering pathway, the metabolic amplifying pathway, and cellular transduction in regulation of glucose-dependent biphasic insulin secretion. Arch Physiol Biochem 2024:1-12. [PMID: 38196246 DOI: 10.1080/13813455.2023.2299920] [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: 08/07/2023] [Accepted: 12/16/2023] [Indexed: 01/11/2024]
Abstract
Introduction: Insulin secretion is a highly regulated process critical for maintaining glucose homeostasis. This abstract explores the intricate interplay between three essential pathways: The Triggering Pathway, The Metabolic Amplifying Pathway, and Cellular Transduction, in orchestrating glucose-dependent biphasic insulin secretion.Mechanism: During the triggering pathway, glucose metabolism in pancreatic beta-cells leads to ATP production, closing ATP-sensitive potassium channels and initiating insulin exocytosis. The metabolic amplifying pathway enhances insulin secretion via key metabolites like NADH and glutamate, enhancing calcium influx and insulin granule exocytosis. Additionally, the cellular transduction pathway involves G-protein coupled receptors and cyclic AMP, modulating insulin secretion.Result and Conclusion: These interconnected pathways ensure a dynamic insulin response to fluctuating glucose levels, with the initial rapid phase and the subsequent sustained phase. Understanding these pathways' complexities provides crucial insights into insulin dysregulation in diabetes and highlights potential therapeutic targets to restore glucose-dependent insulin secretion.
Collapse
Affiliation(s)
- Shradha Bisht
- Amity Institute of Pharmacy, Amity University, Lucknow, Uttar Pradesh, India
| | - Mamta F Singh
- School of Pharmaceutical Sciences, SBS University, Balawala, Uttarakhand, India
| |
Collapse
|
24
|
Mariam Z, Niazi SK. Glucagon-like peptide agonists: A prospective review. Endocrinol Diabetes Metab 2024; 7:e462. [PMID: 38093651 PMCID: PMC10782143 DOI: 10.1002/edm2.462] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/03/2023] [Accepted: 11/19/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have emerged as promising therapeutic options for addressing Type-2 diabetes, obesity, and related conditions. Among these, semaglutide, tirzepatide, liraglutide etc., all notable GLP-1RA, have gained attention owing to their favourable pharmacological properties and clinical efficacy. AIMS This comprehensive review aims to provide a detailed analysis of both the currently available GLP-1RAs in the market and those undergoing clinical trials. The focus is on examining their mechanism of action, pharmacokinetics, efficacy in glycemic control and weight management, safety profile, and potential applications. MATERIALS & METHODS The review employs a systematic approach to gather information on GLP-1RAs. Relevant literature from the currently literature and ongoing clinical trials is thoroughly examined. Detailed scrutiny is applied to understand the mechanism of action, pharmacokinetic properties, and clinical outcomes of these agents. RESULTS The review presents a comprehensive overview of the GLP-1RAs, highlighting their distinct mechanisms of action, pharmacokinetic profiles, and clinical effectiveness in glycemic control and weight management. Safety profiles are also discussed, providing a holistic understanding of these therapeutic agents. DISCUSSION The findings are discussed in the context of advancements in the field of GLP-1RAs. Potential applications beyond diabetes and obesity are explored, shedding light on the broader implications of these agents in managing related conditions. CONCLUSION In conclusion, this review underscores the significance of GLP-1RAs, with a specific focus on semaglutide, in the management of type 2 diabetes, obesity, and beyond. By synthesizing information on their mechanisms, pharmacokinetics, efficacy, and safety, this review provides valuable insights into the potential benefits these agents offer, contributing to the ongoing discourse in the field.
Collapse
|
25
|
Bany Bakar R, Reimann F, Gribble FM. The intestine as an endocrine organ and the role of gut hormones in metabolic regulation. Nat Rev Gastroenterol Hepatol 2023; 20:784-796. [PMID: 37626258 DOI: 10.1038/s41575-023-00830-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
Gut hormones orchestrate pivotal physiological processes in multiple metabolically active tissues, including the pancreas, liver, adipose tissue, gut and central nervous system, making them attractive therapeutic targets in the treatment of obesity and type 2 diabetes mellitus. Most gut hormones are derived from enteroendocrine cells, but bioactive peptides that are derived from other intestinal epithelial cell types have also been implicated in metabolic regulation and can be considered gut hormones. A deeper understanding of the complex inter-organ crosstalk mediated by the intestinal endocrine system is a prerequisite for designing more effective drugs that are based on or target gut hormones and their receptors, and extending their therapeutic potential beyond obesity and diabetes mellitus. In this Review, we present an overview of gut hormones that are involved in the regulation of metabolism and discuss their action in the gastrointestinal system and beyond.
Collapse
Affiliation(s)
- Rula Bany Bakar
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
| |
Collapse
|
26
|
Azizogli AR, Vitti MR, Mishra R, Osorno L, Heffernan C, Kumar VA. Comparison of SGLT1, SGLT2, and Dual Inhibitor biological activity in treating Type 2 Diabetes Mellitus. ADVANCED THERAPEUTICS 2023; 6:2300143. [PMID: 38223846 PMCID: PMC10783160 DOI: 10.1002/adtp.202300143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Indexed: 01/16/2024]
Abstract
Diabetes Mellitus Type 2 (T2D) is an emerging health burden in the USand worldwide, impacting approximately 15% of Americans. Current front-line therapeutics for T2D patients include sulfonylureas that act to reduce A1C and/or fasting blood glucose levels, or Metformin that antagonizes the action of glucagon to reduce hepatic glucose production. Next generation glucomodulatory therapeutics target members of the high-affinity glucose transporter Sodium-Glucose-Linked-Transporter (SGLT) family. SGLT1 is primarily expressed in intestinal epithelium, whose inhibition reduces dietary glucose uptake, whilst SGLT2 is highly expressed in kidney - regulating glucose reabsorption. A number of SGLT2 inhibitors are FDA approved whilst SGLT1 and dual SGLT1 & 2 inhibitor are currently in clinical trials. Here, we discuss and compare SGLT2, SGLT1, and dual inhibitors' biochemical mechanism and physiological effects.
Collapse
Affiliation(s)
- Abdul-Rahman Azizogli
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Michael R Vitti
- University of Virginia School of Medicine, Charlottesville, VA, 22903
| | - Richa Mishra
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Laura Osorno
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Corey Heffernan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, 07103
| |
Collapse
|
27
|
Halkjær SI, de Knegt VE, Kallemose T, Jensen JEB, Cortes D, Gluud LL, Wewer Albrechtsen NJ, Petersen AM. No effect of multi-strain probiotic supplementation on metabolic and inflammatory markers and newborn body composition in pregnant women with obesity: Results from a randomized, double-blind placebo-controlled study. Nutr Metab Cardiovasc Dis 2023; 33:2444-2454. [PMID: 37580231 DOI: 10.1016/j.numecd.2023.07.030] [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: 04/03/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND AND AIMS Modulation of the gut microbiome composition with probiotics may have beneficial metabolic effects in pregnant women with obesity. The aim was to investigate the effect of probiotic supplementation during pregnancy on metabolic and inflammatory markers and the body composition of the offspring. METHODS AND RESULTS A randomized double-blind trial in 50 pregnant women (pre-pregnancy BMI ≥30 and < 35 kg/m2) comparing multi-strain probiotics (Vivomixx®; 450 billion CFU/d) versus placebo from 14 to 20 weeks of gestation until delivery was carried out. Participants were followed with two predelivery visits at gestational week 27-30 and 36-37 and with one postdelivery visit. All visits included fasting blood samples (C-reactive protein (CRP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), insulin, C-peptide, glucose, glucagon, and glucagon-like peptide-1 (GLP-1)). At delivery, umbilical cord blood samples were collected (GLP-1 and glucagon). At the postdelivery visit, a dual-energy X-ray absorptiometry (DXA) scan of the newborn was performed. Forty-nine of 50 participants completed the study until delivery, and 36 mother-offspring dyads underwent postdelivery examinations including a DXA scan. There were no significant differences in changes in measured biomarkers between the probiotic versus the placebo group. No differences were found in newborn body composition or GLP-1 and glucagon. GLP-1 measured in umbilical blood samples was positively correlated to fat percent in offspring from the probiotic group. CONCLUSION In this study of pregnant women with obesity and their newborns, there was no effect of probiotic supplementation in mothers or babies on metabolic or inflammatory biomarkers or on body composition of offspring. This study was registered at clinicaltrials.gov as NCT02508844.
Collapse
Affiliation(s)
- Sofie I Halkjær
- Gastrounit, Medical Section, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Victoria E de Knegt
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thomas Kallemose
- Clinical Research Center, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Jens-Erik B Jensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Dina Cortes
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise L Gluud
- Gastrounit, Medical Section, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Andreas Munk Petersen
- Gastrounit, Medical Section, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Microbiology, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark.
| |
Collapse
|
28
|
Khurana N, Sharma SB. Modulation of glucose metabolism-related genes in diabetic rats treated with herbal synthetic anti-diabetic compound (α-HSA): insights from transcriptomic profiling. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2023; 20:721-728. [PMID: 37401762 DOI: 10.1515/jcim-2023-0156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVES Eugenia jambolana is a medicinal plant traditionally used for treating diabetes. The bioactive compound FIIc, which is derived from the fruit pulp of E. jambolana, has been identified and purified as α-HSA. Previous studies have demonstrated that administration of α-HSA for 6 weeks improved glycemic index and dyslipidemia in rats with T2D. This study investigated the molecular mechanism underlying the potential therapeutic effects of α-HSA in experimentally induced diabetic rats. METHODS Male Wistar rats were divided into four groups: diabetic control, diabetic treated with FIIc, diabetic treated with α-HSA, and diabetic treated with glibenclamide. Over a 6-week experimental period, transcriptomic analysis was conducted on liver, skeletal, and pancreatic tissue samples collected from the rats. RESULTS The study findings revealed significant upregulation of genes associated with glucose metabolism and insulin signaling in the groups treated with FIIc and α-HSA, compared to the diabetic control group. Moreover, pro-inflammatory genes were downregulated in these treatment groups. These results indicate that α-HSA has the potential to modulate key metabolic pathways, improve glucose homeostasis, enhance insulin sensitivity, and alleviate inflammation. CONCLUSIONS This study provides compelling scientific evidence supporting the potential of α-HSA as a therapeutic agent for diabetes treatment. The observed upregulation of genes related to glucose metabolism and insulin signaling, along with the downregulation of pro-inflammatory genes, aligns with the pharmacological activity of α-HSA in controlling glucose homeostasis and improving insulin sensitivity. These findings suggest that α-HSA holds promise as a novel therapeutic approach for managing diabetes and its associated complications.
Collapse
Affiliation(s)
- Nikhil Khurana
- Department of Biochemistry, University College of Medical Sciences and GTB Hospital (University of Delhi), Delhi, India
| | - Suman Bala Sharma
- Departmen of Biochemistry, ESIC Medical College and Hospital, Faridabad, Haryana, India
| |
Collapse
|
29
|
Yang W, Jiang W, Guo S. Regulation of Macronutrients in Insulin Resistance and Glucose Homeostasis during Type 2 Diabetes Mellitus. Nutrients 2023; 15:4671. [PMID: 37960324 PMCID: PMC10647592 DOI: 10.3390/nu15214671] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Insulin resistance is an important feature of metabolic syndrome and a precursor of type 2 diabetes mellitus (T2DM). Overnutrition-induced obesity is a major risk factor for the development of insulin resistance and T2DM. The intake of macronutrients plays a key role in maintaining energy balance. The components of macronutrients distinctly regulate insulin sensitivity and glucose homeostasis. Precisely adjusting the beneficial food compound intake is important for the prevention of insulin resistance and T2DM. Here, we reviewed the effects of different components of macronutrients on insulin sensitivity and their underlying mechanisms, including fructose, dietary fiber, saturated and unsaturated fatty acids, and amino acids. Understanding the diet-gene interaction will help us to better uncover the molecular mechanisms of T2DM and promote the application of precision nutrition in practice by integrating multi-omics analysis.
Collapse
Affiliation(s)
| | | | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA; (W.Y.); (W.J.)
| |
Collapse
|
30
|
Folli F, Finzi G, Manfrini R, Galli A, Casiraghi F, Centofanti L, Berra C, Fiorina P, Davalli A, La Rosa S, Perego C, Higgins PB. Mechanisms of action of incretin receptor based dual- and tri-agonists in pancreatic islets. Am J Physiol Endocrinol Metab 2023; 325:E595-E609. [PMID: 37729025 PMCID: PMC10874655 DOI: 10.1152/ajpendo.00236.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/15/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023]
Abstract
Simultaneous activation of the incretin G-protein-coupled receptors (GPCRs) via unimolecular dual-receptor agonists (UDRA) has emerged as a new therapeutic approach for type 2 diabetes. Recent studies also advocate triple agonism with molecules also capable of binding the glucagon receptor. In this scoping review, we discuss the cellular mechanisms of action (MOA) underlying the actions of these novel and therapeutically important classes of peptide receptor agonists. Clinical efficacy studies of several UDRAs have demonstrated favorable results both as monotherapies and when combined with approved hypoglycemics. Although the additive insulinotropic effects of dual glucagon-like peptide-1 receptor (GLP-1R) and glucose-dependent insulinotropic peptide receptor (GIPR) agonism were anticipated based on the known actions of either glucagon-like peptide-1 (GLP-1) or glucose-dependent insulinotropic peptide (GIP) alone, the additional benefits from GCGR were largely unexpected. Whether additional synergistic or antagonistic interactions among these G-protein receptor signaling pathways arise from simultaneous stimulation is not known. The signaling pathways affected by dual- and tri-agonism require more trenchant investigation before a comprehensive understanding of the cellular MOA. This knowledge will be essential for understanding the chronic efficacy and safety of these treatments.
Collapse
Affiliation(s)
- Franco Folli
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
- Unit of Diabetes, Endocrinology and Metabolism, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Giovanna Finzi
- Unit of Pathology, Department of Oncology, ASST Sette Laghi, Varese, Italy
| | - Roberto Manfrini
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
- Unit of Diabetes, Endocrinology and Metabolism, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Alessandra Galli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Francesca Casiraghi
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Lucia Centofanti
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Cesare Berra
- IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, Milan, Italy
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Alberto Davalli
- Diabetes and Endocrinology Unit, Department of Internal Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano La Rosa
- Unit of Pathology, Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Carla Perego
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Paul B Higgins
- Department of Life & Physical Sciences, Atlantic Technological University, Letterkenny, Ireland
| |
Collapse
|
31
|
Ferguson BK, Wilson PB. Ordered Eating and Its Effects on Various Postprandial Health Markers: A Systematic Review. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2023; 42:746-757. [PMID: 36574255 DOI: 10.1080/27697061.2022.2161664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/18/2022] [Indexed: 10/25/2023]
Abstract
OBJECTIVE Aberrations in glucose, insulin, and other postprandial (PP) markers are common in obesity and cardiometabolic disorders. One potentially simple lifestyle/dietary modification to manage these issues is to change the order in which foods are consumed within meals. Carbohydrate exerts the largest effect on PP glucose, and there is some evidence that ingesting dietary fat or protein before carbohydrate delays gastric emptying of carbohydrate and reduces PP glucose. Additionally, certain dietary proteins may augment insulin release if ingested with carbohydrate, thereby improving blood glucose clearance. This review aimed to systematically evaluate evidence from acute experiments that modified the order in which foods were consumed in isocaloric meals. METHODS Outcomes of interest were PP glucose and insulin (including area under the curve for both), C-peptide, gut hormones, and perceptual responses. Three databases were searched (PubMed, Cochrane CENTRAL, Web of Science) in February 2022. Additionally, reference lists of identified reports were searched, and an author of several studies was consulted to verify that relevant literature was included. The review included acute interventions that administered isocaloric meals of the same foods but with foods eaten in different orders. Studies were not excluded based on participant characteristics. RESULTS Eleven reports were identified. All reports that assessed glucose and insulin showed a tendency toward lower levels, at least over parts of the PP period, by consuming carbohydrates last. GLP-1 tended to be higher in carbohydrate-last conditions, though this was only measured in a few studies. Perceptual responses (hunger, fullness, etc.) were not consistently different between conditions in two studies, but the certainty of evidence was very low. CONCLUSIONS Findings indicate that, at least acutely, there may be benefits to eating carbohydrate after vegetable and/or protein-rich foods. The most consistent effect (judged as moderate certainty) is that carbohydrate-last meal orders tend to lower blood glucose and insulin excursions.
Collapse
Affiliation(s)
- Brian K Ferguson
- Human Performance Laboratory, Human Movement Sciences Department, Old Dominion University, Norfolk, Virginia, USA
| | - Patrick B Wilson
- Human Performance Laboratory, Human Movement Sciences Department, Old Dominion University, Norfolk, Virginia, USA
| |
Collapse
|
32
|
Pusch E, Krążek M, Wojciechowicz T, Sassek M, Kołodziejski PA, Strowski MZ, Nowak KW, Skrzypski M. GIP_HUMAN [22-51] Peptide Encoded by the Glucose-Dependent Insulinotropic Polypeptide (GIP) Gene Suppresses Insulin Expression and Secretion in INS-1E Cells and Rat Pancreatic Islets. Genes (Basel) 2023; 14:1910. [PMID: 37895259 PMCID: PMC10606481 DOI: 10.3390/genes14101910] [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: 09/04/2023] [Revised: 09/24/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
GIP_HUMAN [22-51] is a recently discovered peptide that shares the same precursor molecule with glucose-dependent insulinotropic polypeptide (GIP). In vivo, chronic infusion of GIP_HUMAN [22-51] in ApoE-/- mice enhanced the development of aortic atherosclerotic lesions and upregulated inflammatory and proatherogenic proteins. In the present study, we evaluate the effects of GIP_HUMAN [22-51] on insulin mRNA expression and secretion in insulin-producing INS-1E cells and isolated rat pancreatic islets. Furthermore, we characterize the influence of GIP_HUMAN [22-51] on cell proliferation and death and on Nf-kB nuclear translocation. Rat insulin-producing INS-1E cells and pancreatic islets, isolated from male Wistar rats, were used in this study. Gene expression was evaluated using real-time PCR. Cell proliferation was studied using a BrdU incorporation assay. Cell death was quantified by evaluating histone-complexed DNA fragments. Insulin secretion was determined using an ELISA test. Nf-kB nuclear translocation was detected using immunofluorescence. GIP_HUMAN [22-51] suppressed insulin (Ins1 and Ins2) in INS-1E cells and pancreatic islets. Moreover, GIP_HUMAN [22-51] promoted the translocation of NF-κB from cytoplasm to the nucleus. In the presence of a pharmacological inhibitor of NF-κB, GIP_HUMAN [22-51] was unable to suppress Ins2 mRNA expression. Moreover, GIP_HUMAN [22-51] downregulated insulin secretion at low (2.8 mmol/L) but not high (16.7 mmol/L) glucose concentration. By contrast, GIP_HUMAN [22-51] failed to affect cell proliferation and apoptosis. We conclude that GIP_HUMAN [22-51] suppresses insulin expression and secretion in pancreatic β cells without affecting β cell proliferation or apoptosis. Notably, the effects of GIP_HUMAN [22-51] on insulin secretion are glucose-dependent.
Collapse
Affiliation(s)
- Emily Pusch
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (E.P.); (M.K.); (T.W.); (M.S.); (P.A.K.); (K.W.N.)
| | - Małgorzata Krążek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (E.P.); (M.K.); (T.W.); (M.S.); (P.A.K.); (K.W.N.)
| | - Tatiana Wojciechowicz
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (E.P.); (M.K.); (T.W.); (M.S.); (P.A.K.); (K.W.N.)
| | - Maciej Sassek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (E.P.); (M.K.); (T.W.); (M.S.); (P.A.K.); (K.W.N.)
| | - Paweł A. Kołodziejski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (E.P.); (M.K.); (T.W.); (M.S.); (P.A.K.); (K.W.N.)
| | - Mathias Z. Strowski
- Department of Hepatology and Gastroenterology, Charité-University Medicine Berlin, 13353 Berlin, Germany;
- Medical Clinic III, 15236 Frankfurt, Germany
| | - Krzysztof W. Nowak
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (E.P.); (M.K.); (T.W.); (M.S.); (P.A.K.); (K.W.N.)
| | - Marek Skrzypski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, 60-637 Poznan, Poland; (E.P.); (M.K.); (T.W.); (M.S.); (P.A.K.); (K.W.N.)
| |
Collapse
|
33
|
Safarzad M, Jazi MS, Kiaei M, Asadi J. Lower serum zinc level is associated with higher fasting insulin in type 2 diabetes mellitus (T2DM) and relates with disturbed glucagon suppression response in male patients. Prim Care Diabetes 2023; 17:493-498. [PMID: 37391316 DOI: 10.1016/j.pcd.2023.05.008] [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: 02/17/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 07/02/2023]
Abstract
AIMS Zinc ion can play critical role in glycemic control in diabetes mellitus (DM), contributing to both insulin synthesis and secretion. In this study, we aimed to investigate the level of zinc in diabetic patients and its association with glycemic parameters, insulin, and glucagon level. METHODS 112 individuals (59 cases of type 2DM and 53 non-diabetic controls) were included in this study. Biochemical parameters (FBG, 2hpp, HbA1C), and zinc level in the serum were measured using colorimetric assays. Insulin and glucagon were measured by ELISA method. HOMA-IR, HOMA-B, reciprocal HOMA-B, and Quicki indices were calculated using appropriate formula. For further analysis, patients were divided into two groups: high (>135.5 μg/dl) and low (<135.5 μg/dl) zinc. Glucagon suppression was considered yes if 2hpp glucagon < fasting glucagon. RESULTS Our results showed that serum Zn level in type 2 DM patients was lower than control (P value=0.02). Patients with lower Zn had higher fasting insulin (P value=0.006) and higher β-cell activity index (HOMA-B, p value=0.02), however fasting glucagon and parameters of hyperglycemia (FBG, 2hpp, Hba1C) were not different. Moreover, insulin sensitivity and resistance indices (Quicki, HOMA-IR,1/HOMA-IR) showed non-significantly improved status in high Zn group. We found non-significant association between glucagon suppression and Zn level in both genders (N = 39, p value = 0.07), however, it was significant in males (N = 14, p value = 0.02). CONCLUSION Altogether, our results indicated reduced serum Zn in type 2DM can exacerbate hyperinsulinemia and glucagon suppression (only significant in the male), highlighting its importance in type 2DM control.
Collapse
Affiliation(s)
- Mahdieh Safarzad
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Marie Saghaeian Jazi
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Mohammadreza Kiaei
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Jahanbakhsh Asadi
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
34
|
Ahmad BA, Sanghani IM, Sayabugari R, Biju H, Siddegowda A, Ittiachen Kinattingal M, Yartha SGR, Gaonkar PM, Andrabi SS, Vaghamashi YK, Korwar A. Beyond Blood Sugar: Investigating the Cardiovascular Effects of Antidiabetic Drugs. Cureus 2023; 15:e46373. [PMID: 37920618 PMCID: PMC10618835 DOI: 10.7759/cureus.46373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2023] [Indexed: 11/04/2023] Open
Abstract
Cardiovascular disease is a major comorbidity associated with diabetes mellitus. Various antidiabetic drugs are currently used to treat type 2 diabetes mellitus and have varying effects on the cardiovascular system. Some drugs, such as glucagon-like peptide 1 (GLP-1) agonists and sodium-glucose cotransporter 2 (SGLT-2) inhibitors, are cardioprotective, whereas others, such as insulin, have deleterious effects on the cardiovascular system. This narrative review assessed the impact of antidiabetic drugs on cardiovascular health in the management of diabetes mellitus. It critically examines various classes of these medications, including conventional options such as metformin and newer agents such as incretin-based therapies and SGLT-2.
Collapse
Affiliation(s)
- Binish A Ahmad
- Department of Internal Medicine, King Edward Medical University, Lahore, PAK
| | - Isha M Sanghani
- Department of Internal Medicine, Punyashlok Ahilyadevi Holkar Government Medical College, Baramati, IND
| | | | - Hannah Biju
- Department of Internal Medicine, Kristu Jayanti College, Bengaluru, IND
| | | | - Minnu Ittiachen Kinattingal
- Department of Internal Medicine, New Hope Clinical Research, Charlotte, USA
- Department of Internal Medicine, Karuna Medical College, Palakkad, IND
| | | | - Prajyoth M Gaonkar
- Department of Internal Medicine, Punyashlok Ahilyadevi Holkar Government Medical College, Baramati, IND
| | - Syed Shireen Andrabi
- Department of Internal Medicine, School of Medicne, Tehran University of Medical Sciences, Tehran, IRN
| | | | - Arunika Korwar
- Department of Internal Medicine, KJ Somaiya Medical College, Mumbai, IND
| |
Collapse
|
35
|
Freitas FPC, Rodrigues CEM. Effect of liraglutide on cardiometabolic profile and on bioelectrical impedance analysis in patients with obesity and metabolic syndrome. Sci Rep 2023; 13:13090. [PMID: 37567946 PMCID: PMC10421848 DOI: 10.1038/s41598-023-40366-4] [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: 04/14/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023] Open
Abstract
Metabolic syndrome (MetS) and obesity represent a public health problem worldwide. Bioelectrical impedance analysis (BIA) is a practical and effective way of evaluating body composition, especially abdominal fat. Liraglutide, a GLP-1 analog, reduces body weight and improves cardiometabolic parameters. In this prospective non-randomized intervention study, we evaluated the effect of 6 months of treatment with liraglutide (n = 57) on the clinical, laboratory and BIA findings of adult sex-stratified patients diagnosed with obesity and MetS, compared to a control group receiving sibutramine (n = 46). The groups were statistically similar with regard to the age of females (p = 0.852) and males (p = 0.657). Almost all anthropometric and BIA variables were higher in the treatment group than in the comparative group (p < 0.05). Abdominal circumference (AC) decreased significantly more in the treatment group. In males, body weight and fat mass also decreased (p < 0.05). Liraglutide treatment was associated with a greater reduction in trunk fat mass (FMT) (p < 0.05). AC and FMT were strongly correlated (rho = 0.531, p < 0.001) in the treatment group. In the multiple regression analysis, liraglutide treatment remained independently associated with FMT. Treatment with liraglutide for 6 months promoted weight loss, improved cardiometabolic and inflammatory parameters and led to a significant reduction in FMT correlated with AC in obese MetS patients of both sexes.
Collapse
Affiliation(s)
| | - Carlos Ewerton Maia Rodrigues
- Graduate Program in Medical Sciences, Universidade de Fortaleza (Unifor), Fortaleza, Brazil.
- Department of Internal Medicine, Federal University of Ceará, Rua Gontran Giffoni 366, Apto 301, Torre I, Patriolino Ribeiro, Fortaleza, Ceará, CEP 60810-220, Brazil.
| |
Collapse
|
36
|
Chen L, Yun Y, Guo S, Wang X, Xiong M, Zhao T, Xu T, Shen J, Xie X, Wang K. Discovery of Novel 5,6-Dihydro-1,2,4-triazine Derivatives as Efficacious Glucagon-Like Peptide-1 Receptor Agonists. J Med Chem 2023. [PMID: 37286364 DOI: 10.1021/acs.jmedchem.3c00320] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Danuglipron is the most representative small-molecule agonist of the glucagon-like peptide-1 receptor (GLP-1R) and has received considerable attention due to positive results in the treatment of type 2 diabetes mellitus (T2DM) and obesity in clinical trials. However, hERG inhibition, lower activity than endogenous GLP-1, and a short action time represent limitations in terms of feasible application. In this study, we report a new class of 5,6-dihydro-1,2,4-triazine derivatives that serve to eliminate potential hERG inhibition caused by the piperidine ring of danuglipron. Applying systematic in vitro to in vivo screening, we have identified compound 42 as a highly potent and selective GLP-1R agonist, which delivers improved (7-fold) efficacy in stimulating cAMP accumulation compared with danuglipron and which exhibits acceptable drug-like properties. Furthermore, 42 significantly reduces glucose excursion and inhibits food intake of hGLP-1R Knock-In mice. These effects are longer-lasting than that shown by danuglipron, demonstrating feasibility in the treatment of T2DM and obesity.
Collapse
Affiliation(s)
- Lili Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Ying Yun
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Shimeng Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoyan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Muya Xiong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Tingting Zhao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Tifei Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianhua Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xin Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
37
|
Pandey S, Mangmool S, Parichatikanond W. Multifaceted Roles of GLP-1 and Its Analogs: A Review on Molecular Mechanisms with a Cardiotherapeutic Perspective. Pharmaceuticals (Basel) 2023; 16:836. [PMID: 37375783 DOI: 10.3390/ph16060836] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetes is one of the chronic metabolic disorders which poses a multitude of life-debilitating challenges, including cardiac muscle impairment, which eventually results in heart failure. The incretin hormone glucagon-like peptide-1 (GLP-1) has gained distinct recognition in reinstating glucose homeostasis in diabetes, while it is now largely accepted that it has an array of biological effects in the body. Several lines of evidence have revealed that GLP-1 and its analogs possess cardioprotective effects by various mechanisms related to cardiac contractility, myocardial glucose uptake, cardiac oxidative stress and ischemia/reperfusion injury, and mitochondrial homeostasis. Upon binding to GLP-1 receptor (GLP-1R), GLP-1 and its analogs exert their effects via adenylyl cyclase-mediated cAMP elevation and subsequent activation of cAMP-dependent protein kinase(s) which stimulates the insulin release in conjunction with enhanced Ca2+ and ATP levels. Recent findings have suggested additional downstream molecular pathways stirred by long-term exposure of GLP-1 analogs, which pave the way for the development of potential therapeutic molecules with longer lasting beneficial effects against diabetic cardiomyopathies. This review provides a comprehensive overview of the recent advances in the understanding of the GLP-1R-dependent and -independent actions of GLP-1 and its analogs in the protection against cardiomyopathies.
Collapse
Affiliation(s)
- Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | |
Collapse
|
38
|
Coulter-Parkhill A, Dobbin S, Tanday N, Gault VA, McClean S, Irwin N. A novel peptide isolated from Aphonopelma chalcodes tarantula venom with benefits on pancreatic islet function and appetite control. Biochem Pharmacol 2023; 212:115544. [PMID: 37044298 DOI: 10.1016/j.bcp.2023.115544] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Proof-of-concept for therapeutic application of venom-derived compounds in diabetes is exemplified by the incretin mimetic, exenatide, originally extracted from the saliva of the venomous Heloderma suspectum lizard. In this regard, we have isolated and sequenced a novel 28 amino acid peptide named Δ-theraphotoxin-Ac1 (Δ-TRTX-AC1) from venom of the Mexican Blond tarantula spider Aphonopelma chalcodes, with potential therapeutic benefits for diabetes. Following confirmation of the structure and safety profile of the synthetic peptide, assessment of enzymatic stability and effects of Δ-TRTX-AC1 on in vitro beta-cell function were studied, alongside potential mechanisms. Glucose homeostatic and satiety actions of Δ-TRTX-AC1 alone, and in combination with exenatide, were then assessed in C57BL/6 mice. Synthetic Δ-TRTX-AC1 was shown to adopt a characteristic inhibitor cysteine knot (ICK)-like structure and was non-toxic to beta-cells. Δ-TRTX-AC1 evoked glucose-dependent insulin secretion from BRIN BD11 cells with bioactivity confirmed in murine islets. Insulin secretory potency was established to be dependent on KATP and Ca2+ channel beta-cell signalling. In addition, Δ-TRTX-AC1 enhanced beta-cell proliferation and provided significant protection against cytokine-induced apoptosis. When injected co-jointly with glucose in mice at a dose of 250 nmol/kg, Δ-TRTX-AC1 decreased blood-glucose levels and evoked a significant satiating effect. Moreover, whilst Δ-TRTX-AC1 did not enhance exenatide induced benefits on glucose homeostasis, the peptide significantly augmented exenatide mediated suppression of appetite. Together these data highlight the therapeutic potential of tarantula spider venom-derived peptides, such as Δ-TRTX-Ac1, for diabetes and related obesity.
Collapse
Affiliation(s)
| | - Swm Dobbin
- Ulster University, Coleraine, Northern Ireland, UK
| | - N Tanday
- Ulster University, Coleraine, Northern Ireland, UK
| | - V A Gault
- Ulster University, Coleraine, Northern Ireland, UK
| | - S McClean
- Ulster University, Coleraine, Northern Ireland, UK
| | - N Irwin
- Ulster University, Coleraine, Northern Ireland, UK.
| |
Collapse
|
39
|
Hall LG, Thyfault JP, Johnson JD. Exercise and inactivity as modifiers of β cell function and type 2 diabetes risk. J Appl Physiol (1985) 2023; 134:823-839. [PMID: 36759159 PMCID: PMC10042613 DOI: 10.1152/japplphysiol.00472.2022] [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: 08/15/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Exercise and regular physical activity are beneficial for the prevention and management of metabolic diseases such as obesity and type 2 diabetes, whereas exercise cessation, defined as deconditioning from regular exercise or physical activity that has lasted for a period of months to years, can lead to metabolic derangements that drive disease. Adaptations to the insulin-secreting pancreatic β-cells are an important benefit of exercise, whereas less is known about how exercise cessation affects these cells. Our aim is to review the impact that exercise and exercise cessation have on β-cell function, with a focus on the evidence from studies examining glucose-stimulated insulin secretion (GSIS) using gold-standard techniques. Potential mechanisms by which the β-cell adapts to exercise, including exerkine and incretin signaling, autonomic nervous system signaling, and changes in insulin clearance, will also be explored. We will highlight areas for future research.
Collapse
Affiliation(s)
- Liam G Hall
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
- KU Diabetes Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
40
|
Ajabnoor GMA, Hashim KT, Alzahrani MM, Alsuheili AZ, Alharbi AF, Alhozali AM, Enani S, Eldakhakhny B, Elsamanoudy A. The Possible Effect of the Long-Term Use of Glucagon-like Peptide-1 Receptor Agonists (GLP-1RA) on Hba1c and Lipid Profile in Type 2 Diabetes Mellitus: A Retrospective Study in KAUH, Jeddah, Saudi Arabia. Diseases 2023; 11:diseases11010050. [PMID: 36975599 PMCID: PMC10046996 DOI: 10.3390/diseases11010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
(1) Background: Type 2 diabetes (T2DM) is a chronic metabolic disease with serious health complications. T2DM is associated with many chronic illnesses, including kidney failure, cardiovascular diseases (CVD), vision loss, and other related diseases. Obesity is one of the major factors associated with insulin resistance and dyslipidemia. Recently, the development of GLP-1 Receptor agonist (GLP-1RA) showed great therapeutic potential for T2DM. Aim: To retrospectively investigate the association of the long-term use of GLP-1RA therapy in T2DM patients with HbA1c levels and dyslipidemia. (2) Methods: Retrospective data collection and analysis of demographic, clinical records, and biochemical parameters were carried out for 72 T2DM taking GLP-1RA treatments for six months. (3) Results: A total of 72 T2DM patients with a mean age = 55 (28 male and 44 female) were divided into two groups. Group 1 received statins (n = 63), and group 2 did not receive statins (n = 9). The GLP-1RA effect on BMI was significantly decreased in group 1 (p < 0.01). A significant effect was observed for HbA1c in both groups for six months of treatment duration (p < 0.05). The AST levels significantly decreased in group 2 from 25.2 to 19.4 U\L (p = 0.011). (4) Conclusions: GLP-1RA treatments were associated with weight reduction and improved glycemic control for T2DM patients. Moreover, it is suggested that it has anti-inflammatory and hepatoprotective effects. However, no direct association was found with the lipid profile in all groups of T2DM.
Collapse
Affiliation(s)
- Ghada M. A. Ajabnoor
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- King Abdulaziz University Hospital, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Kamal Talat Hashim
- Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | | | | | | | - Amani Matook Alhozali
- King Abdulaziz University Hospital, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- Department of Medicine, College of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Sumia Enani
- Department of Food and Nutrition, Faculty of Human Sciences and Design, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Basmah Eldakhakhny
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Ayman Elsamanoudy
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Correspondence:
| |
Collapse
|
41
|
Lee SH, Ko HM, Jee W, Kim H, Chung WS, Jang HJ. Isosinensetin Stimulates Glucagon-like Peptide-1 Secretion via Activation of hTAS2R50 and the G βγ-Mediated Signaling Pathway. Int J Mol Sci 2023; 24:ijms24043682. [PMID: 36835092 PMCID: PMC9959872 DOI: 10.3390/ijms24043682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Bitter taste receptors (TAS2Rs) are G protein-coupled receptors localized in the taste buds of the tongue. They may also be present in non-lingual organs, including the brain, lung, kidney, and gastrointestinal (GI) tract. Recent studies on bitter taste receptor functions have suggested TAS2Rs as potential therapeutic targets. The human bitter taste receptor subtype hTAS2R50 responds to its agonist isosinensetin (ISS). Here, we demonstrated that, unlike other TAS2R agonists, isosinensetin activated hTAS2R50 as well as increased Glucagon-like peptide 1 (GLP-1) secretion through the Gβγ-mediated pathway in NCI-H716 cells. To confirm this mechanism, we showed that ISS increased intracellular Ca2+ and was suppressed by the IP3R inhibitor 2-APB as well as the PLC inhibitor U73122, suggesting that TAS2Rs alters the physiological state of enteroendocrine L cells in a PLC-dependent manner. Furthermore, we demonstrated that ISS upregulated proglucagon mRNA and stimulated GLP-1 secretion. ISS-mediated GLP-1 secretion was suppressed in response to small interfering RNA-mediated silencing of Gα-gust and hTAS2R50 as well as 2-APB and U73122. Our findings improved the understanding of how ISS modulates GLP-1 secretion and indicates the possibility of using ISS as a therapeutic agent in the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Seung-Hyeon Lee
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyun Min Ko
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wona Jee
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyungsuk Kim
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Won-Seok Chung
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hyeung-Jin Jang
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence:
| |
Collapse
|
42
|
Diabetic Kidney Disease in Post-Transplant Diabetes Mellitus: Causes, Treatment and Outcomes. Biomedicines 2023; 11:biomedicines11020470. [PMID: 36831005 PMCID: PMC9953284 DOI: 10.3390/biomedicines11020470] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 02/08/2023] Open
Abstract
Kidney transplant recipients are a unique subgroup of chronic kidney disease patients due to their single functioning kidney, immunosuppressive agent usage, and long-term complications related to transplantation. Post-transplant diabetes mellitus (PTDM) has a significant adverse effect on renal outcomes in particular. As transplantations enable people to live longer, cardiovascular morbidity and mortality become more prevalent, and PTDM is a key risk factor for these complications. Although PTDM results from similar risk factors to those of type 2 diabetes, the conditions differ in their pathophysiology and clinical features. Transplantation itself is a risk factor for diabetes due to chronic exposure to immunosuppressive agents. Considering current evidence, this article describes the risk factors, pathogenesis, diagnostic criteria, prevention strategies, and management of PTDM. The therapeutic options are discussed regarding their safety and potential drug-drug interactions with immunosuppressive agents.
Collapse
|
43
|
Hori M, Hasegawa Y, Hayashi Y, Nakagami T, Harada-Shiba M. Acute Cholesterol-Lowering Effect of Exendin-4 in Ldlr -/- and C57BL/6J Mice. J Atheroscler Thromb 2023; 30:74-86. [PMID: 35314564 PMCID: PMC9899697 DOI: 10.5551/jat.60921] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIMS We previously reported that glucagon-like peptide-1 receptor agonists (GLP-1RAs) reduced serum low-density lipoprotein cholesterol (LDL-C) levels in patients with type 2 diabetes mellitus receiving statins, which increased LDL receptor (LDLR) expression. Nevertheless, it remains unclear how much LDLR expression contributes to the LDL-C-lowering effect of GLP-1RAs. We examined the effect of a GLP-1RA, namely, exendin-4, on serum LDL-C levels and its mechanism in Ldlr-/- and C57BL/6J mice. METHODS Ten-week-old Ldlr-/- and C57BL/6J mice received exendin-4 or saline for 5 days, and serum lipid profiles and hepatic lipid levels were examined. Cholesterol metabolism-related gene expression and protein levels in the liver and ileum and the fecal bile acid (BA) composition were also examined. RESULTS Exendin-4 treatment significantly decreased serum very-low-density lipoprotein cholesterol (VLDL-C) and LDL-C levels and mature hepatic SREBP2 levels and increased hepatic Insig1/2 mRNA expression in both mouse strains. In Ldlr-/- mice, exendin-4 treatment also significantly decreased hepatic cholesterol levels and fecal BA excretion, decreased hepatic Cyp7a1 mRNA expression, and increased small intestinal Fgf15 mRNA expression. In C57BL/6J mice, exendin-4 treatment significantly decreased small intestinal NPC1L1 levels. CONCLUSIONS Our findings demonstrate that exendin-4 treatment decreased serum VLDL-C and LDL-C levels in a manner that was independent of LDLR. Exendin-4 treatment might decrease serum cholesterol levels by lowering hepatic SREBP2 levels and cholesterol absorption in Ldlr-/- and C57BL/6J mice. Exendin-4 treatment might decrease cholesterol absorption by different mechanisms in Ldlr-/- and C57BL/6J mice.
Collapse
Affiliation(s)
- Mika Hori
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan,Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan,Department of Endocrinology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Yukiko Hasegawa
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan,Division of Diabetology and Metabolism, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Yoshitaka Hayashi
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan,Department of Endocrinology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Tomoko Nakagami
- Division of Diabetology and Metabolism, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| |
Collapse
|
44
|
Unlu Y, Vinales KL, Hollstein T, Chang D, Cabeza de Baca T, Walter M, Krakoff J, Piaggi P. The association between gut hormones and diet-induced metabolic flexibility in metabolically healthy adults. Obesity (Silver Spring) 2023; 31:139-149. [PMID: 36471908 PMCID: PMC9780166 DOI: 10.1002/oby.23584] [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: 05/12/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study investigated whether interindividual variance in diet-induced metabolic flexibility is explained by differences in gut hormone concentrations. METHODS A total of 69 healthy volunteers with normal glucose regulation underwent 24-hour assessments of respiratory quotient (RQ) in a whole-room indirect calorimeter during eucaloric feeding (EBL; 50% carbohydrate, 30% fat) and then, in a crossover design, during 24-hour fasting and three normal-protein (20%) overfeeding diets (200% energy requirements). Metabolic flexibility was defined as the change in 24-hour RQ from EBL during standard (50% carbohydrate), high-fat (60%), and high-carbohydrate (75%) overfeeding diets. Plasma concentrations of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) after an overnight fast were measured prior to and after each diet. RESULTS Compared with EBL, on average, 24-hour RQ decreased by ~4% during high-fat overfeeding, whereas it increased by ~4% during standard overfeeding and by ~9% during high-carbohydrate overfeeding. During high-carbohydrate overfeeding, but not during any other overfeeding diet or fasting, increased GLP-1 concentration was associated with increased RQ (r = 0.44, p < 0.001), higher/lower carbohydrate/lipid oxidation rates (r = 0.34 and r = -0.51, both p < 0.01), respectively, and increased plasma insulin concentration (r = 0.38, p = 0.02). CONCLUSIONS Increased GLP-1 concentration following high-carbohydrate overfeeding associated with a greater shift to carbohydrate oxidation, suggesting that GLP-1 may be implicated in diet-induced metabolic flexibility to carbohydrate overload.
Collapse
Affiliation(s)
- Yigit Unlu
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
| | - Karyne L. Vinales
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
- Endocrinology Division, Medicine Department, Phoenix VA Health Care System, Phoenix, AZ, USA
| | - Tim Hollstein
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
- Division of Endocrinology, Diabetology and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Arnold-Heller-Straße 3, Kiel 24105, Germany
| | - Douglas Chang
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
| | - Tomás Cabeza de Baca
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
| | - Mary Walter
- Clinical Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
| | - Paolo Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ
- Department of Information Engineering, University of Pisa, Pisa, Italy
| |
Collapse
|
45
|
Mirzaei F, Khodadadi I, Majdoub N, Vafaei SA, Tayebinia H, Abbasi E. Role of Glucagon-like Peptide-1 (GLP-1) Agonists in the Management of Diabetic Patients with or without COVID-19. THE OPEN MEDICINAL CHEMISTRY JOURNAL 2022. [DOI: 10.2174/18741045-v16-e2212130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) is a gut-derived hormone released after a meal, which alleviates hyperglycemia, increases β-cell survival, reduces body weight, and reduces inflammation. These thrilling effects motivated clinical studies to discover the potential use of GLP-1 receptor agonists (GLP-1 RAs) in the management of T2D. GLP-1 RAs are potential anti-diabetic agents that can reduce blood pressure, glucose levels, HbA1c and, weight loss without hypoglycemia risk. This manuscript reviews the importance of GLP-1 RAs and their role in the management of T2D with or without COVID-19 infection. Hence, this manuscript can help physicians and researchers to choose the most appropriate drugs for the individualized treatment of subjects.
Collapse
|
46
|
Baafi K, March JC. Harnessing gut cells for functional insulin production: Strategies and challenges. BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2022; 4:7-13. [PMID: 39416909 PMCID: PMC11446352 DOI: 10.1016/j.biotno.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 10/19/2024]
Abstract
Reprogrammed glucose-responsive, insulin + cells ("β-like") exhibit the potential to bypass the hurdles of exogenous insulin delivery in treating diabetes mellitus. Current cell-based therapies-transcription factor regulation, biomolecule-mediated enteric signaling, and transgenics - have demonstrated the promise of reprogramming either mature or progenitor gut cells into surrogate "β-like" cells. However, there are predominant challenges impeding the use of gut "β-like" cells as clinical replacements for insulin therapy. Reprogrammed "β-like" gut cells, even those of enteroendocrine origin, mostly do not exhibit glucose - potentiated insulin secretion. Despite the exceptionally low conversion rate of gut cells into surrogate "β-like" cells, the therapeutic quantity of gut "β-like" cells needed for normoglycemia has not even been established. There is also a lingering uncertainty regarding the functionality and bioavailability of gut derived insulin. Herein, we review the strategies, challenges, and opportunities in the generation of functional, reprogrammed "β-like" cells.
Collapse
Affiliation(s)
- Kelvin Baafi
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | | |
Collapse
|
47
|
Nugraheni M, Purwanti S, Ekawatiningsih P. Chemical composition, glycaemic index, and antidiabetic property of analogue rice made from composite tubers, germinated legumes, and cereal flours. INTERNATIONAL FOOD RESEARCH JOURNAL 2022. [DOI: 10.47836/ifrj.29.6.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The dependence on rice as a source of carbohydrates in Indonesia is among the highest in Asia. Innovations to develop products that can be used as a carbohydrate source, and have functional values beneficial to health are currently needed. The present work thus aimed to determine the chemical composition, glycaemic index, and antidiabetic property of three analogue rice types. The formulation of three types of analogue rice was done by combining natural tuber flour, modified tuber flour, germinated cereals, and germinated legumes. The glycaemic index was assessed using experimental animal. The antidiabetic properties of three types of analogue rice were assessed by food efficiency ratio, glucose profile, lipid profile, and atherogenic index. Results showed that analogue rice had high dietary fibre, resistant starch, and protein, and low fat and carbohydrate. The three types of analogue rice were classified as low glycaemic index based on glycaemic response tests. The glycaemic index of analogue rice I, II, and III were 41.23 ± 3.39, 42.55 ± 3.21, and 40.19 ± 3.34, respectively. The ability of analogue rice to decrease glucose, triglycerides, total cholesterol, low-density lipoprotein, atherogenic index; and increase high-density lipoprotein in diabetic mice was affected by its low glycaemic index and chemical composition benefits. The ability to improve the characteristics of glucose and lipids should support the development of analogue rice as a functional food.
Collapse
|
48
|
The Interaction of Food Allergy and Diabetes: Food Allergy Effects on Diabetic Mice by Intestinal Barrier Destruction and Glucagon-like Peptide 1 Reduction in Jejunum. Foods 2022; 11:foods11233758. [PMID: 36496564 PMCID: PMC9741085 DOI: 10.3390/foods11233758] [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: 09/30/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
The increase in food allergies and diabetes leads to the assumption that they are related. This study aimed to (1) verify the interaction between food allergy and diabetes and (2) explore the potential mechanisms by which food allergy promotes diabetes. Female BALB/c mice were grouped into a control group (CK), an ovalbumin-sensitized group (OVA), a diabetes group (STZ), and a diabetic allergic group (STZ + OVA) (Mice were modeled diabetes with STZ first, then were given OVA to model food allergies), and an allergic diabetic group (OVA + STZ) (Mice were modeled food allergies with OVA first, then were given STZ to model diabetes). The results showed that OVA + STZ mice exhibited a more serious Th2 humoral response, and they were more susceptible to diabetes. Furthermore, when the OVA + STZ mice were in the sensitized state, the intestinal barrier function was severely impaired, and mast cell activation was promoted. Moreover, we found that the effect of food allergy on diabetes is related to the inhibition of GLP-1 secretion and the up-regulation of the PI3K/Akt/mTOR/NF-κB P65 signaling pathway in the jejunum. Overall, our results suggest that food allergies have interactions with diabetes, which sheds new light on the importance of food allergies in diabetes.
Collapse
|
49
|
Park SE, Kang S, Paek J, Georgescu A, Chang J, Yi AY, Wilkins BJ, Karakasheva TA, Hamilton KE, Huh DD. Geometric engineering of organoid culture for enhanced organogenesis in a dish. Nat Methods 2022; 19:1449-1460. [PMID: 36280722 PMCID: PMC10027401 DOI: 10.1038/s41592-022-01643-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/09/2022] [Indexed: 12/22/2022]
Abstract
Here, we introduce a facile, scalable engineering approach to enable long-term development and maturation of organoids. We have redesigned the configuration of conventional organoid culture to develop a platform that converts single injections of stem cell suspensions to radial arrays of organoids that can be maintained for extended periods without the need for passaging. Using this system, we demonstrate accelerated production of intestinal organoids with significantly enhanced structural and functional maturity, and their continuous development for over 4 weeks. Furthermore, we present a patient-derived organoid model of inflammatory bowel disease (IBD) and its interrogation using single-cell RNA sequencing to demonstrate its ability to reproduce key pathological features of IBD. Finally, we describe the extension of our approach to engineer vascularized, perfusable human enteroids, which can be used to model innate immune responses in IBD. This work provides an immediately deployable platform technology toward engineering more realistic organ-like structures in a dish.
Collapse
Affiliation(s)
- Sunghee Estelle Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Shawn Kang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jungwook Paek
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrei Georgescu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Vivodyne, Inc., Philadelphia, PA, USA
| | - Jeehan Chang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex Yoon Yi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin J Wilkins
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tatiana A Karakasheva
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathryn E Hamilton
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Madsen CT, Refsgaard JC, Teufel FG, Kjærulff SK, Wang Z, Meng G, Jessen C, Heljo P, Jiang Q, Zhao X, Wu B, Zhou X, Tang Y, Jeppesen JF, Kelstrup CD, Buckley ST, Tullin S, Nygaard-Jensen J, Chen X, Zhang F, Olsen JV, Han D, Grønborg M, de Lichtenberg U. Combining mass spectrometry and machine learning to discover bioactive peptides. Nat Commun 2022; 13:6235. [PMID: 36266275 PMCID: PMC9584923 DOI: 10.1038/s41467-022-34031-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 10/10/2022] [Indexed: 12/25/2022] Open
Abstract
Peptides play important roles in regulating biological processes and form the basis of a multiplicity of therapeutic drugs. To date, only about 300 peptides in human have confirmed bioactivity, although tens of thousands have been reported in the literature. The majority of these are inactive degradation products of endogenous proteins and peptides, presenting a needle-in-a-haystack problem of identifying the most promising candidate peptides from large-scale peptidomics experiments to test for bioactivity. To address this challenge, we conducted a comprehensive analysis of the mammalian peptidome across seven tissues in four different mouse strains and used the data to train a machine learning model that predicts hundreds of peptide candidates based on patterns in the mass spectrometry data. We provide in silico validation examples and experimental confirmation of bioactivity for two peptides, demonstrating the utility of this resource for discovering lead peptides for further characterization and therapeutic development.
Collapse
Affiliation(s)
| | - Jan C Refsgaard
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
- Intomics, Kongens Lyngby, Denmark
| | - Felix G Teufel
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
| | - Sonny K Kjærulff
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
- Intomics, Kongens Lyngby, Denmark
| | - Zhe Wang
- Novo Nordisk Research Centre China, Beijing, China
| | - Guangjun Meng
- Novo Nordisk Research Centre China, Beijing, China
- Pulmongene LTD. Rm 502, Building 2, No. 9, Yike Road, Zhongguancun Life Science Park, Changping District, Beijing, China
| | - Carsten Jessen
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
| | - Petteri Heljo
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
| | - Qunfeng Jiang
- Novo Nordisk Research Centre China, Beijing, China
- Innovent Biologics, Inc. DongPing Jie 168, Suzhou, China
| | - Xin Zhao
- Novo Nordisk Research Centre China, Beijing, China
| | - Bo Wu
- Novo Nordisk Research Centre China, Beijing, China
- QL Biopharmaceutical, Rm 101, Building 7, 20 Life Science Park Road, Beijing, China
| | - Xueping Zhou
- Novo Nordisk Research Centre China, Beijing, China
- Crinetics pharmaceuticals, 10222 Barnes Canyon Rd Building 2, San Diego, CA, 92121, USA
| | - Yang Tang
- Novo Nordisk Research Centre China, Beijing, China
- Roche R&D Center (China) Ltd, Building 5, 371 Lishizhen Road, 201203, Pudong, Shanghai, China
| | - Jacob F Jeppesen
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
| | | | | | - Søren Tullin
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
- Boehringer Ingelheim GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach, Germany
| | - Jan Nygaard-Jensen
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
- Boehringer Ingelheim GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach, Germany
| | - Xiaoli Chen
- Novo Nordisk Research Centre China, Beijing, China
| | - Fang Zhang
- Novo Nordisk Research Centre China, Beijing, China
- Structure Therapeutics. 701 Gateway Blvd., South San Francisco, CA, 94080, USA
| | - Jesper V Olsen
- Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Dan Han
- Novo Nordisk Research Centre China, Beijing, China
| | - Mads Grønborg
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
| | - Ulrik de Lichtenberg
- Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
- The Novo Nordisk Foundation, Tuborg Havnevej 19, 2900, Hellerup, Denmark
| |
Collapse
|