Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 28, 2024; 30(36): 4036-4043
Published online Sep 28, 2024. doi: 10.3748/wjg.v30.i36.4036
Glucagon-like peptide-1 receptor agonists: Exploring the mechanisms from glycemic control to treatment of multisystemic diseases
Mo-Wei Kong, Yang Yu, Department of Cardiology, Southwest Medical University, Luzhou 646000, Sichuan Province, China
Ying Wan, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, China
Yu Gao, Department of Endocrinology, The Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, China
Chun-Xiang Zhang, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
Chun-Xiang Zhang, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou 646000, Sichuan Province, China
ORCID number: Mo-Wei Kong (0000-0002-1214-164X); Chun-Xiang Zhang (0000-0001-8303-6495).
Co-first authors: Mo-Wei Kong and Yang Yu.
Author contributions: Kong MW and Yu Y wrote the manuscript. Designation of Kong MW and Yu Y as joint first authors are based on three main reasons. First, the research was conducted as a collaborative effort, and the designation of joint first authorship accurately reflects the distribution of responsibilities and burdens associated with the time and effort required to complete the study and the subsequent paper. This also ensures effective communication and management of post-submission matters, ultimately enhancing the quality and reliability of the paper. Second, the entire research team comprises authors with diverse expertise and skills from various fields, and the designation of joint first authors best represent this diversity. This facilitates the most comprehensive and in-depth examination of the research topic, enriching readers' understanding by offering multiple expert perspectives. Third, Kong MW and Yu Y contributed efforts of equal substance throughout the research process. Selecting these researchers as joint first authors acknowledge and respect this equal contribution, while recognizing the spirit of teamwork and collaboration of this study. Zhang CX provided crucial suggestions and guidance for the writing; Wan Y and Gao Y reviewed and revised the manuscript; all authors read and approved the final manuscript.
Supported by National Natural Science Foundation of China, No. U23A20398 and No. 82030007; Sichuan Science and Technology Program, No. 2022YFS0578.
Conflict-of-interest statement: The authors declare that they have no competing interests.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Chun-Xiang Zhang, MD, Dean, Professor, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, No. 1735 West Harrison Street, Luzhou 646000, Sichuan Province, China. zhangchx999@163.com
Received: July 27, 2024
Revised: August 8, 2024
Accepted: September 5, 2024
Published online: September 28, 2024
Processing time: 54 Days and 13.3 Hours

Abstract

This editorial takes a deeper look at the insights provided by Soresi and Giannitrapani, which examined the therapeutic potential of glucagon-like peptide-1 receptor agonists (GLP-1RAs) for metabolic dysfunction-associated fatty liver disease. We provide supplementary insights to their research, highlighting the broader systemic implications of GLP-1RAs, synthesizing the current understanding of their mechanisms and the trajectory of research in this field. GLP-1RAs are revolutionizing the treatment of type 2 diabetes mellitus and beyond. Beyond glycemic control, GLP-1RAs demonstrate cardiovascular and renal protective effects, offering potential in managing diabetic kidney disease al-ongside renin–angiotensin–aldosterone system inhibitors. Their role in bone metabolism hints at benefits for diabetic osteoporosis, while the neuroprotective properties of GLP-1RAs show promise in Alzheimer's disease treatment by modulating neuronal insulin signaling. Additionally, they improve hormonal and metabolic profiles in polycystic ovary syndrome. This editorial highlights the multifaceted mechanisms of GLP-1RAs, emphasizing the need for ongoing research to fully realize their therapeutic potential across a range of multisystemic diseases.

Key Words: Glucagon-like peptide-1 receptor agonists; Glycemic control; Multisystem diseases; Mechanism of action; Cardiovascular protection; Renal disease; Bone metabolism; Non-alcoholic fatty liver disease; Neuroprotection; Polycystic ovary syndrome

Core Tip: This editorial serves as an extension to the insightful research by Soresi and Giannitrapani, initially presented in the World Journal of Gastroenterology. We revisit their focus on the role of glucagon-like peptide-1 receptor agonists (GLP-1RAs) in treating metabolic dysfunction-associated fatty liver disease, and supplement their work by exploring the broader systemic applications of GLP-1RAs. Our aim is to provide a comprehensive overview of the mechanisms of action of GLP-1RAs and to highlight the latest research developments, emphasizing their potential in managing a variety of diseases. We emphasize the multifaceted therapeutic value of GLP-1RAs and encourage further investigation into their clinical utility across different medical specialties.



INTRODUCTION

This editorial reviews the potential role of glucagon-like peptide-1 receptor agonists (GLP-1RAs) in the treatment of metabolic dysfunction-associated fatty liver disease, as discussed by Soresi and Giannitrapani[1] in World Journal of Gastroenterology. GLP-1RAs show promise in reducing fat and body weight, and improving insulin resistance (IR), which may play a role in protecting the liver from further damage. Although clinical trials have shown that GLP-1RAs, such as semaglutide, can reduce fatty liver, including histological signs of nonalcoholic steatohepatitis, their effects on fibrosis are limited. Therefore, combination therapies with different but complementary mechanisms of action are considered the best way to improve efficacy and slow disease progression. We demonstrate that although international drug agencies have not yet given clear indications, more treatment plans involving GLP-1RA combination therapies may emerge in the future.

The mechanism by which GLP-1RAs improve the sugar and fat metabolism of skeletal muscle cells was important in our previous research research[2,3]. Glucagon-like peptide (GLP) originates from the L cells of the intestine and exerts its effects by binding to glucagon-like peptide-1 receptor (GLP-1R). GLP-1 reduces glucagon secretion and plays a blood-sugar-lowering role by increasing insulin secretion dependent on postprandial glucose concentration. GLP-1RAs, with longer half-life and dipeptidyl peptidase-4 resistance, mimic the effects of GLP-1 and are extensively used in diabetes treatment[4]. Currently, the main GLP-1RAs on the market include liraglutide, exenatide and dulaglutide. Recent research has found that GLP-1RAs promote insulin secretion and regulate blood sugar, have cardiovascular benefits, kidney protection, and improve fatty liver, and cognitive function[5]. This editorial focuses on exploring the potential of GLP-1 analogs in the treatment of various diseases.

IMPACT OF GLP-1RAS ON THE CARDIOVASCULAR SYSTEM

In the cardiovascular system, GLP-1Rs are expressed in both myocardial and endothelial cells[6], indicating that GLP-1Rs have direct and indirect effects on the heart and blood vessels (Figure 1). A multicenter long-term cardiovascular outcome trial confirmed that GLP-1RAs can reduce the mortality of cardiovascular diseases and are beneficial to the cardiovascular system, reducing the incidence of myocardial infarction or nonfatal stroke[7]. GLP-1RAs may improve endothelial cell function through anti-inflammatory and vasodilatory activities. The recent STEP-HFpEF trial[8] found that semaglutide significantly reduced heart-failure-related symptoms in patients with obesity-related preserved ejection fraction heart failure and type 2 diabetes mellitus (T2DM) within 1 year. In addition, the antiproliferative effect on smooth muscle cells and the anti-inflammatory effect on macrophages help prevent the occurrence and progression of atherosclerosis. Some researchers have proposed that GLP-1RAs can enhance insulin sensitivity in myocardial cells, increase the rate of glucose oxidation, reduce the rate of fatty acid oxidation, and enhance the energy metabolism of myocardial cells, thereby improving heart function[9]. An animal experiment of ischemia–reperfusion injury showed that early treatment with natural GLP-1 and GLP-1RAs can increase myocardial blood flow, reduce the area of myocardial infarction, and improve myocardial injury after ischemia–reperfusion. Cardiac calcium metabolism disorder is a sign of heart failure, and studies have shown that GLP-1RAs can regulate the downregulation of calcium ion levels caused by reduced phosphorylation of the ryanodine receptor and activation of calmodulin-dependent protein kinase II[10]. GLP-1RAs can improve myocardial remodeling by activating the NO synthase/cyclic GMP/cyclic GMP-dependent protein kinase signal transduction pathway. Although clinical studies have confirmed some cardiovascular protective effects of GLP-1RAs in diabetic patients, more basic research is needed to support the molecular mechanisms by which GLP-1RAs exert their cardiovascular protective effects.

Figure 1
Figure 1 Glucagon-like peptide-1 receptors are expressed in both cardiomyocytes and endothelial cells, exerting direct and indirect effects on the heart and blood vessels. GLP-1: Glucagon-like peptide-1; GLP-1R: Glucagon-like peptide-1 receptor; LV: Left ventricle; TV: Tricuspid valve.
IMPACT OF GLP-1RAS ON BONE METABOLISM

Diabetes and osteoporosis are closely linked, with diabetes leading to fragile bones, increased fracture risk, and slow healing due to metabolic disorders caused by high blood sugar. Some blood-sugar-lowering medications, such as thiazolidinediones and insulin, may increase the risk of fractures by affecting bone metabolism[11]. Therefore, diabetes treatment strategies need to effectively control blood sugar while minimizing impacts on bone metabolic complications. GLP-1RAs can exert antiosteoporotic effects by controlling blood sugar levels, promoting bone formation, and inhibiting bone resorption, and primarily enhance bone formation through physiological effects that stimulate insulin secretion, inhibit glucagon secretion, and regulate gastric emptying[12]. GLP-1RAs may beneficially affect the skeleton of diabetic patients by interacting with the advanced glycation end products (AGEs)-RAGE-ROS pathway[13]. Thus, GLP-1RAs hold potential value in the treatment of diabetes and associated osteoporosis. Animal studies have shown that liraglutide can significantly reduce AGE and reduce bone absorption in a model of chronic diabetic bone injury complications in ApoE gene knockout mice[14,15]. However, the specific impact of GLP-1RAs on fracture risk and osteoporosis has not yet been clearly defined. Some studies have shown that GLP-1RAs have a good anabolic effect on bone metabolism, but the potential molecular mechanisms still need to be clarified[16].

IMPACT OF GLP-1RAS ON POLYCYSTIC OVARY SYNDROME

Polycystic ovary syndrome (PCOS), characterized by infrequent or absent ovulation, elevated androgen levels, polycystic ovaries, obesity, and infertility, is the most common endocrine and metabolic disorder in women of reproductive age[17]. Although the pathogenesis of PCOS is not fully understood, recent studies suggest a causal relationship between IR and all clinical symptoms of PCOS. The IR in PCOS disrupts the action of gonadotropins, enhances the sensitivity of theca cells to luteinizing hormone, leading to increased androgen production[18]. Concurrently, elevated insulin levels reduce the binding of sex-hormone-binding globulin in the liver to testosterone, resulting in hyperandrogenism[19]. High levels of free fatty acids, a primary manifestation of obesity, can exacerbate IR by activating serine/threonine kinases, which reduce the tyrosine phosphorylation of insulin receptor substrate-1[20]. Weight loss has been shown to improve reproductive function, hyperandrogenemia, and metabolism in women with PCOS. GLP-1RA, known for its weight loss and IR improvement effects, has been widely used in the treatment of PCOS. GLP-1RAs can reduce body weight in PCOS patients and lower androgen levels. However, research on the underlying mechanisms of GLP-1RAs in PCOS treatment has been limited. The proliferation and apoptosis of granulosa cells are fundamental to follicular development and atresia, with the forkhead box transcription factor O1 playing a crucial role in promoting PCOS follicular atresia and granulosa cell apoptosis[21]. Recent studies have found that the gut microbe Bacteroides vulgatus, through its metabolite difluoro methylarginine (DFMA), activates the farnesoid X receptor pathway, which subsequently suppresses GLP-1 secretion. This suppression leads to IR and ovarian dysfunction. However, the GLP-1RA liraglutide and the arginine decarboxylase inhibitor DFMA have been shown to improve ovarian dysfunction in mouse models that mimic PCOS[22], thereby presenting new potential targets for the treatment of PCOS (Figure 2). Other research has found that liraglutide can repair cognitive impairments in PCOS rat models by inhibiting overexpression of the Notch signaling pathway[23]. Therefore, based on preliminary research and clinical efficacy assessments, GLP-1RA has shown promising therapeutic effects in the treatment of PCOS.

Figure 2
Figure 2 Glucagon-like peptide-1 receptor agonists improve ovarian dysfunction in a mouse model of polycystic ovary syndrome. GLP-1RA: Glucagon-like peptide-1 receptor agonists; GLP-1: Glucagon-like peptide-1; PCOS: Polycystic ovary syndrome; DFMA: Difluoro methylarginine.
GLP-1RA AND ALZHEIMER'S DISEASE

Alzheimer's disease (AD) is the most common neurodegenerative disease. Despite extensive research over the past few decades, the mechanisms of occurrence and development of AD have not been fully elucidated. Autopsy analysis has revealed that in the brains of AD patients with significantly reduced insulin receptor expression, IR also occurs as the disease progresses, indicating that defects in insulin signaling are related to the pathogenesis of AD[24]. Epidemiological studies have found a correlation between T2DM and AD[25]. Experimental studies have shown that most GLP-1 analogs can cross the blood–brain barrier and have a direct physiological effect on the human brain[26]. These effects include increasing the proliferation of neural progenitor cells, improving learning, and reducing the formation of plaques and inflammation in the brain. Experimental studies have also confirmed that GLP-1RAs can reduce the levels of AD pathological markers, such as oligomeric Aβ and Aβ plaque burden, reduce the activation of microglia, and improve memory[27]. Overexpression of GLP-1Rs in the hippocampus of mice has been found to increase neurite growth and improve spatial learning[28]. GLP-1RAs can reduce cell necrosis caused by glutamate, Fe2+ ions, and hypoxia, thereby protecting hippocampal neurons[29]. Calcium also plays an important role in neuronal plasticity and neurodegenerative diseases. In vitro experiments have found that pretreatment of rat hippocampal neurons with GLP-1 can weaken the response of Ca2+ ions to glutamate and weaken membrane depolarization, reducing the susceptibility of neurons to glutamate-induced death[30]. Studies have found that dulaglutide can protect learning and memory, reduce the hyperphosphorylation of tau protein, and its mechanism may be related to the phosphatidylinositol 3-kinase/serine-threonine kinase/glycogen synthase kinase-3β signaling pathway[30,31]. Based on the above analysis, although more clinical evidence is needed, the existing data has shown the potential application prospects and great research potential of GLP-1 and its analogs in the future treatment of AD.

IMPACT OF GLP-1RAS ON THE KIDNEYS

Diabetic kidney disease (DKD) is one of the most important microvascular complications of diabetes and is one of the important causes of chronic and end-stage kidney diseases. Its clinical manifestations are proteinuria and a decline in glomerular filtration rate. Even a slight increase in proteinuria or a decline in glomerular filtration rate is significantly related to an increased risk of cardiovascular diseases and cardiovascular death, so kidney protection is an important goal in the treatment of T2DM[32]. In addition to renin-angiotensin-aldosterone system (RAAS) inhibitors, there are currently no other drugs available for the treatment of DKD. GLP-1RAs have been proven to prevent the occurrence of proteinuria in diabetic patients and reduce the decline in glomerular filtration rate[33]. In humans, GLP-1R is present in cells of the proximal renal tubules and preglomerular vascular smooth muscle cells[34]. The FLOW trial demonstrated that semaglutide treatment significantly reduced the risk of major kidney disease events and cardiovascular death in patients with T2DM and chronic kidney disease. Studies have confirmed that GLP-1 can induce urinary sodium excretion and diuresis, and its mechanism may be to inhibit the sodium–hydrogen exchange protein located on the brush border of cells in the proximal renal tubules[35]. It rapidly downregulates the activity of Na+/K+-ATPase distributed on the brush border surface of the renal proximal tubules, causing pressure diuresis, which can partially explain the antihypertensive effect of GLP-1RAs[36]. The activation of the intrarenal RAAS is a well-known pathogenic medium of DKD. In experimental studies, GLP-1 and GLP-1RAs have been proven to reduce the concentration of angiotensin II. GLP-1 promotes the excretion of water and sodium by inhibiting the release of aldosterone and may also reduce hypertension, mesangial cell injury, and renal fibrosis induced by angiotensin II, thereby playing a role in protecting the kidneys[37]. GLP-1 reduces the production of reactive oxygen species in diabetic kidneys by activating the cyclic AMP-protein kinase A signal transduction pathway, inhibits oxidative stress, and protects the kidneys from oxidative damage. GLP-1RAs can significantly inhibit the activity of nuclear factor-κB in kidney tissue and significantly reduce expression of CD14, intercellular adhesion molecule-1, transforming growth factor-β1, and type IV collagen protein levels, indicating that GLP-1RAs can play a protective role in DKD by inhibiting the inflammatory response, and this effect is independent of the hypoglycemic, antihypertensive, and weight reduction effects[38-40]. The above data lay the foundation for future clinical research on whether GLP-1RAs can improve the prognosis of DKD patients.

CONCLUSION

GLP-1RAs are innovative medications for treating T2DM, offering benefits such as reduced fasting glucose, lowered glycated hemoglobin, decreased blood pressure, and induced weight loss; all with a minimal risk of hypoglycemia. Their multifaceted action includes neuroprotection and cardiovascular and metabolic regulation, which emphasizes their potential in managing a variety of neurological and endocrine disorders. Despite ongoing debates about their tolerability and side effects, ongoing research is exploring oral formulations and enhancers to optimize their therapeutic profile. This editorial systematically reviews the broader impacts of GLP-1RAs, beyond glycemic control, and their therapeutic potential across different diseases (Figure 3), aiming to pave the way for future investigative directions.

Figure 3
Figure 3 Glucagon-like peptide-1 receptor agonists are extensively involved in various systemic diseases and hold therapeutic potential for a range of conditions. GLP-1R: Glucagon-like peptide-1 receptor; GLP-1: Glucagon-like peptide-1; GI: Gastrointestinal.
Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade C

Creativity or Innovation: Grade C

Scientific Significance: Grade B

P-Reviewer: Li SY S-Editor: Lin C L-Editor: A P-Editor: Cai YX

References
1.  Soresi M, Giannitrapani L. Glucagon-like peptide 1 agonists are potentially useful drugs for treating metabolic dysfunction-associated steatotic liver disease. World J Gastroenterol. 2024;30:3541-3547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
2.  Kong MW, Gao Y, Xie YY, Xing EH, Sun LX, Ma HJ, Xing HY. Mechanism of GLP-1 Receptor Agonists-Mediated Attenuation of Palmitic Acid-Induced Lipotoxicity in L6 Myoblasts. Biomed Res Int. 2022;2022:6237405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
3.  Tian X, Gao Y, Kong M, Zhao L, Xing E, Sun Q, He J, Lu Y, Feng Z. GLP‑1 receptor agonist protects palmitate-induced insulin resistance in skeletal muscle cells by up-regulating sestrin2 to promote autophagy. Sci Rep. 2023;13:9446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
4.  Zheng K, Azhie A, You X, Naghibzadeh M, Tan E, Naimimohasses S, Sridhar VS, Gupta S, Chen S, Dash S, Tsien C, Selzner N, Lilly L, Jaeckel E, Woo M, Singh S, Cherney D, Bhat M. Glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter-2 inhibitors for the treatment of diabetes mellitus in liver transplant recipients. Diabetes Obes Metab. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
5.  Pechmann LM, Pinheiro FI, Andrade VFC, Moreira CA. The multiple actions of dipeptidyl peptidase 4 (DPP-4) and its pharmacological inhibition on bone metabolism: a review. Diabetol Metab Syndr. 2024;16:175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
6.  Zhao X, Zeng Q, Yu S, Zhu X, Bin Hu, Deng L, Zhang Y, Liu Y. GLP-1R mediates idebenone-reduced blood glucose in mice. Biomed Pharmacother. 2024;178:117202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
7.  He X, Wen S, Tang X, Wen Z, Zhang R, Li S, Gao R, Wang J, Zhu Y, Fang D, Li T, Peng R, Zhang Z, Wen S, Zhou L, Ai H, Lu Y, Zhang S, Shi G, Chen Y. Glucagon-like peptide-1 receptor agonists rescued diabetic vascular endothelial damage through suppression of aberrant STING signaling. Acta Pharm Sin B. 2024;14:2613-2630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
8.  Apperloo EM, Neuen BL, Fletcher RA, Jongs N, Anker SD, Bhatt DL, Butler J, Cherney DZI, Herrington WG, Inzucchi SE, Jardine MJ, Liu CC, Mahaffey KW, McGuire DK, McMurray JJV, Neal B, Packer M, Perkovic V, Sabatine MS, Solomon SD, Staplin N, Szarek M, Vaduganathan M, Wanner C, Wheeler DC, Wiviott SD, Zannad F, Heerspink HJL. Efficacy and safety of SGLT2 inhibitors with and without glucagon-like peptide 1 receptor agonists: a SMART-C collaborative meta-analysis of randomised controlled trials. Lancet Diabetes Endocrinol. 2024;12:545-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
9.  Kosiborod MN, Petrie MC, Borlaug BA, Butler J, Davies MJ, Hovingh GK, Kitzman DW, Møller DV, Treppendahl MB, Verma S, Jensen TJ, Liisberg K, Lindegaard ML, Abhayaratna W, Ahmed FZ, Ben-Gal T, Chopra V, Ezekowitz JA, Fu M, Ito H, Lelonek M, Melenovský V, Merkely B, Núñez J, Perna E, Schou M, Senni M, Sharma K, van der Meer P, Von Lewinski D, Wolf D, Shah SJ; STEP-HFpEF DM Trial Committees and Investigators. Semaglutide in Patients with Obesity-Related Heart Failure and Type 2 Diabetes. N Engl J Med. 2024;390:1394-1407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Reference Citation Analysis (0)]
10.  Huang XD, Jiang DS, Feng X, Fang ZM. The benefits of oral glucose-lowering agents: GLP-1 receptor agonists, DPP-4 and SGLT-2 inhibitors on myocardial ischaemia/reperfusion injury. Eur J Pharmacol. 2024;976:176698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Abdin A, Böhm M, Shahim B, Karlström P, Kulenthiran S, Skouri H, Lund LH. Heart failure with preserved ejection fraction epidemiology, pathophysiology, diagnosis and treatment strategies. Int J Cardiol. 2024;412:132304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
12.  Liu S, Anderson PJ, Rajagopal S, Lefkowitz RJ, Rockman HA. G Protein-Coupled Receptors: A Century of Research and Discovery. Circ Res. 2024;135:174-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
13.  Cui C, Wu SL, Chen JJ, Gongpan P, Guan M, Geng CA. Sesquiterpenoids from Alpinia oxyphylla with GLP-1 Stimulative Effects through Ca(2+)/CaMKII and PKA Pathways and Multiple-Enzyme Inhibition. J Agric Food Chem. 2023;71:16148-16159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
14.  Wang N, Lin Z, Gao L, Wang B, Wei K, Zhang M, Li Y, Xue P. Liraglutide reduces bone marrow adipogenesis by miR-150-5p/ GDF11 axis in diabetic rats. Eur J Pharmacol. 2024;978:176793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
15.  Wu Z, Deng W, Ye Y, Xu J, Han D, Zheng Y, Zheng Q. Liraglutide, a glucagon-like peptide-1 receptor agonist, inhibits bone loss in an animal model of osteoporosis with or without diabetes. Front Endocrinol (Lausanne). 2024;15:1378291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
16.  Al Refaie A, Baldassini L, Mondillo C, Ceccarelli E, Tarquini R, Gennari L, Gonnelli S, Caffarelli C. Glucagon-like Peptide-1 Receptor Agonists and Diabetic Osteopathy: Another Positive Effect of Incretines? A 12 Months Longitudinal Study. Calcif Tissue Int. 2024;115:160-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
17.  Nowicki JK, Jakubowska-Pietkiewicz E. Osteocalcin: Beyond Bones. Endocrinol Metab (Seoul). 2024;39:399-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
18.  Zhang Y, Wang S, Zhou Q, Xie Y, Hu Y, Fang W, Yang C, Wang Z, Ye S, Wang X, Zheng C. Novel Angiogenesis Role of GLP-1(32-36) to Rescue Diabetic Ischemic Lower Limbs via GLP-1R-Dependent Glycolysis in Mice. Arterioscler Thromb Vasc Biol. 2024;44:1225-1245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
19.  Nassar M, Misra A, Bloomgarden Z. Impact of treatment with GLP-1RAs on suicide attempts in adults persons with type 2 diabetes: A retrospective comparative effectiveness study based on a global TriNetX health research database. J Diabetes. 2024;16:e13547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
20.  Arvanitakis K, Chatzikalil E, Kalopitas G, Patoulias D, Popovic DS, Metallidis S, Kotsa K, Germanidis G, Koufakis T. Metabolic Dysfunction-Associated Steatotic Liver Disease and Polycystic Ovary Syndrome: A Complex Interplay. J Clin Med. 2024;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
21.  Gan J, Chen J, Ma RL, Deng Y, Ding XS, Zhu SY, Sun AJ. Action Mechanisms of Metformin Combined with Exenatide and Metformin Only in the Treatment of PCOS in Obese Patients. Int J Endocrinol. 2023;2023:4288004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
22.  Singh V, Mahra K, Jung D, Shin JH. Gut Microbes in Polycystic Ovary Syndrome and Associated Comorbidities; Type 2 Diabetes, Non-Alcoholic Fatty Liver Disease (NAFLD), Cardiovascular Disease (CVD), and the Potential of Microbial Therapeutics. Probiotics Antimicrob Proteins. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
23.  Bao X, Yan D, Yang J, Zhang Z, Yuan B. Role of ERβ in the ovary and ovary related diseases. Gene. 2024;927:148678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Reference Citation Analysis (0)]
24.  Yun C, Yan S, Liao B, Ding Y, Qi X, Zhao M, Wang K, Zhuo Y, Nie Q, Ye C, Xia P, Ma M, Li R, Jiang C, Qiao J, Pang Y. The microbial metabolite agmatine acts as an FXR agonist to promote polycystic ovary syndrome in female mice. Nat Metab. 2024;6:947-962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
25.  Koike H, Harada M, Kusamoto A, Kunitomi C, Xu Z, Tanaka T, Urata Y, Nose E, Takahashi N, Wada-Hiraike O, Hirota Y, Koga K, Osuga Y. Notch Signaling Induced by Endoplasmic Reticulum Stress Regulates Cumulus-Oocyte Complex Expansion in Polycystic Ovary Syndrome. Biomolecules. 2022;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
26.  Zamora MG, García-Lluch G, Moreno L, Pardo J, Pericas CC. Assessment of sodium-glucose cotransporter 2 inhibitors (SGLT2i) and other antidiabetic agents in Alzheimer's disease: A population-based study. Pharmacol Res. 2024;206:107295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Drucker DJ. Efficacy and Safety of GLP-1 Medicines for Type 2 Diabetes and Obesity. Diabetes Care. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Liu M, Ma N, Yang X, Sun M, Li X, Liu Y, Chang Q, Hei C. The Association of Circulating Glucagon-Like Peptide-1 with Cognitive Functions and Biomarkers in Alzheimer's Disease. J Alzheimers Dis. 2024;99:525-533.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
29.  Zhang M, Wu Y, Gao R, Chen X, Chen R, Chen Z. Glucagon-like peptide-1 analogs mitigate neuroinflammation in Alzheimer's disease by suppressing NLRP2 activation in astrocytes. Mol Cell Endocrinol. 2022;542:111529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 16]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
30.  Yu HY, Sun T, Wang Z, Li H, Xu D, An J, Wen LL, Li JY, Li W, Feng J. Exendin-4 and linagliptin attenuate neuroinflammation in a mouse model of Parkinson's disease. Neural Regen Res. 2023;18:1818-1826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
31.  de Paiva IHR, da Silva RS, Mendonça IP, de Souza JRB, Peixoto CA. Semaglutide Attenuates Anxious and Depressive-Like Behaviors and Reverses the Cognitive Impairment in a Type 2 Diabetes Mellitus Mouse Model Via the Microbiota-Gut-Brain Axis. J Neuroimmune Pharmacol. 2024;19:36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
32.  Shao YQ, Wang YC, Wang L, Ruan HZ, Liu YF, Zhang TH, Weng SJ, Yang XL, Zhong YM. Topical administration of GLP-1 eyedrops improves retinal ganglion cell function by facilitating presynaptic GABA release in early experimental diabetes. Neural Regen Res. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
33.  El Mahdy RN, Nader MA, Helal MG, Abu-Risha SE, Abdelmageed ME. Tiron ameliorates acetic acid-induced colitis in rats: Role of TGF-β/EGFR/PI3K/NF-κB signaling pathway. Int Immunopharmacol. 2024;128:111587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
34.  Wajdlich M, Nowicki M. The impact of GLP-1 receptor agonist liraglutide on blood pressure profile, hydration, natriuresis in diabetic patients with severely impaired kidney function. Sci Rep. 2024;14: 5002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
35.  Scurt FG, Ganz MJ, Herzog C, Bose K, Mertens PR, Chatzikyrkou C. Association of metabolic syndrome and chronic kidney disease. Obes Rev. 2024;25:e13649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
36.  Hu Q, Chen Y, Deng X, Li Y, Ma X, Zeng J, Zhao Y. Diabetic nephropathy: Focusing on pathological signals, clinical treatment, and dietary regulation. Biomed Pharmacother. 2023;159:114252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 42]  [Reference Citation Analysis (0)]
37.  Botfield HF, Uldall MS, Westgate CSJ, Mitchell JL, Hagen SM, Gonzalez AM, Hodson DJ, Jensen RH, Sinclair AJ. A glucagon-like peptide-1 receptor agonist reduces intracranial pressure in a rat model of hydrocephalus. Sci Transl Med. 2017;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 58]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
38.  Gao S, Li Y, Liu MM, Xiong X, Cui CP, Huo QJ, Li KX, Sun X, Zhang R, Wu D, Li BY. The crucial relationship between miRNA-27 and CSE/H(2)S, and the mechanism of action of GLP-1 in myocardial hypertrophy. Int J Med Sci. 2024;21:965-977.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
39.  Singh AK, Farag YMK, Zheng Z, Bakris GL. Clinical trial designs of emerging therapies for diabetic kidney disease (DKD). Postgrad Med. 2024;24:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
40.  Bonnet F, Cooper ME, Kopp L, Fouque D, Candido R. A review of the latest real-world evidence studies in diabetic kidney disease: What have we learned about clinical practice and the clinical effectiveness of interventions? Diabetes Obes Metab. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]