Next Article in Journal
Annexin A5 Inhibits Endothelial Inflammation Induced by Lipopolysaccharide-Activated Platelets and Microvesicles via Phosphatidylserine Binding
Previous Article in Journal
Synthesis, In Silico Studies, and Antioxidant and Tyrosinase Inhibitory Potential of 2-(Substituted Phenyl) Thiazolidine-4-Carboxamide Derivatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multifaceted Roles of GLP-1 and Its Analogs: A Review on Molecular Mechanisms with a Cardiotherapeutic Perspective

by
Sudhir Pandey
1,
Supachoke Mangmool
2 and
Warisara Parichatikanond
1,*
1
Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
2
Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(6), 836; https://doi.org/10.3390/ph16060836
Submission received: 10 May 2023 / Revised: 1 June 2023 / Accepted: 1 June 2023 / Published: 3 June 2023
(This article belongs to the Section Pharmacology)

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.

1. Introduction

Metabolic diseases have been on the rise globally, and novel therapeutics with lesser or no side effects to encounter the unmet medical needs of new-age morbid situations are highly needed [1,2]. From that perspective, growth factor signaling, particularly insulin signaling, is one of the most demanding pathways for cell growth, metabolism, and homeostasis. Nevertheless, any deviation or interruption of its normal processes could lead to the development of cardiometabolic disorders and premature death or senescence of the cells [3].
Glucagon-like peptide 1 (GLP-1) is an incretin peptide hormone that aids in insulin secretion, thereby contributing to the regulation and homeostasis of bioenergetic pathway and cell survival. In the human body, GLP-1 is mainly secreted by enteroendocrine L cells in the distal intestine, pancreatic α-cells, and the central nervous system (CNS) [4]. After a meal, GLP-1 is immediately released, leading to an increase in glucose-stimulated insulin secretion (GSIS) at physiological plasma levels. Previous studies suggested that factors from the gut provoke the pancreas to aid in nutrient assimilation, which laid the foundation for the treatment of diabetic patients using extracts from porcine small intestine [5]. The first incretin hormone, gastric inhibitory polypeptide (GIP), isolated from crude extracts of the porcine small intestine, inhibited gastric acid secretion [6]. Later, it was found that GIP stimulated insulin secretion in both animals and humans; it was renamed as glucose-dependent insulinotropic polypeptide; however, the acronym was remained as the same [7].
The second incretin hormone, GLP-1, was discovered in the pancreatic islets from anglerfish. GLP-1 and GLP-2 were identified in hamsters and humans; however, GLP-1 is the main isoform that could stimulate insulin secretion upon diet ingestion [8,9]. GLP-1 is produced in two bioactive forms consisting of glycine-extended GLP-1(7-37) and amidated GLP-1(7-36) via the post-translational processing of proglucagon by proprotein convertase subtilisin-kexin type 1 (PCSK1) or type 3 (PCSK3) [10,11] (Figure 1). GLP-1 receptor (GLP-1R) is classified in class B of the G protein-coupled receptor (GPCR) family that is abundant in the CNS and pancreas, while it is also found in lower expression in the heart, lungs, gut, muscles, kidneys, liver, peripheral nervous system (PNS), and other tissues [3]. GLP-1R gene expressions (transcripts per million, TPM) amongst females and males in the cardiovascular system and pancreas (from GTEx Portal https://gtexportal.org/home/gene/GLP1R accessed on 21 November 2022) are shown in Figure 2. Upon binding to GLP-1R, GLP-1 and its analogs initiate a series of events, including the activation of membrane-bound adenylyl cyclase (AC) and the subsequent production of cyclic adenosine monophosphate (cAMP). Several signal transduction pathways can be initiated downstream of cAMP formation via its effectors such as protein kinase A (PKA) and exchange protein activated by cAMP (Epac) [12,13].
GLP-1(7-36) has a relatively short half-life, which is cleaved by the activity of enzyme dipeptidyl peptidase-4 (DPP-4) at its N-terminus to produce GLP-1(9-36). It was previously supposed that GLP-1(9-36) is an inactive GLP-1 metabolite due to its poor interaction with GLP-1R and lack of significant effects on stimulating insulin secretion or glucose homeostasis [14]. However, recent studies showed that GLP-1(9-36) exerts cardioprotective effects against oxidative injury in H9c2 cardiomyoblasts through the phosphoinositide 3-kinase (PI3K)/Akt/nitric oxide synthase (NOS) pathway in an GLP-1R independent mechanism [15]. Similar cardioprotective roles of GLP-1(9-36) have been observed in dog models with dilated cardiomyopathy [16] and cultured cardiac cells of Goto–Kakizaki rats [17]. It has been suggested that GLP-1(9-36) amide is an active form at physiological concentration, while other GLP-1 metabolites, such as GLP-1(28-36) and GLP-1(32-36), are metabolically inactive [18]. However, growing evidence suggested that some of these metabolites may contribute to pleiotropic effects of GLP-1, irrespective of the GLP-1R [19,20]. At pharmacological doses, these metabolites possess glucoregulatory and cytoprotective effects such as reducing oxidative stress in vascular tissues [21], protecting pancreatic β-cells [22], and suppressing oxidative stress and gluconeogenesis in liver [23].
After a meal, GLP-1 is secreted from the L-cells in a biphasic pattern that is an early immediate rise of GLP-1′s plasma concentration starting approximately 10 to 15 min after ingestion, followed by a late increase achieving the maximum level at 30–60 min, and then a gradual decline to baseline over several hours [24]. However, due to local GLP-1 degradation by DPP-4 and further degradation by DPP-4 in the liver, only 10–15% of endogenously released GLP-1 reaches the systemic circulation [25]. Due to the rapid degradation of GLP-1 by DPP-4, exogenously administered GLP-1 has a short half-life of 1–2 min. Therefore, several GLP-1 analogs (or GLP-1 mimetics) have been developed in two ways, GLP-1R agonists resistant to DPP-4-mediated degradation and DPP-4 inhibitors, both of which tend to prolong the retention time of circulating GLP-1(7-36) amide [26]. To avoid degradation by DPP-4, GLP-1 analogs have been developed by modifying the amino acid residues in the native GLP-1. Additionally, they were synthetically created through the modifications of the structure of exendin-4, a 39-amino-acid peptide initially obtained from the saliva of the lizard Heloderma suspectum, which activated GLP-1R and is inherently resistant to DPP-4 degradation [27,28].
GLP-1 analogs such as liraglutide and exendin-4 are not rapidly degraded by DPP-4, allowing them to exert long therapeutic effects that would be impossible with endogenous GLP-1 due to their extremely short half-life in systemic circulation [29,30]. GLP-1 analogs exert a plethora of additional beneficial effects beyond antidiabetic effects, including increased satiety, decreased gastric emptying, replenishment of insulin stores, cytoprotective properties, and anti-inflammatory effects [31,32,33]. Currently, there are various GLP-1 analogs approved for the treatment of type 2 diabetes (T2DM) which were developed from human GLP-1 backbone (_glutide) (e.g., albiglutide, dulaglutide, liraglutide, and semaglutide) and exendin-4 backbone (_natide) (e.g., exenatide, efpeglenatide, and lixisenatide) (Table 1). These drugs are considered more efficient than DPP-4 inhibitors that block the breakdown of GLP-1; instead, they directly activate the GLP-1R signaling [34,35].

2. GLP-1 Analogs and Physiological Importance in Humans

GLP-1 and its analogs have emerged as attractive approaches to target the secondary manifestations due to hyperglycemic or diabetic conditions in the human body. Beyond their antidiabetic effects, GLP-1 analogs not only reduce diet-induced pancreatic secretions, but also diminish gastric emptying [40]. The ileal-brake activity of GLP-1 was confirmed using the GLP-1R antagonist, exendin-(9-39), which was suggested to have physiologic significance [41,42]. The inhibitory action of GLP-1 was later discovered to be mediated by a vagal pathway [43]. The systemic effects of GLP-1 and its analogs on various vital tissues and organs include reduction of insulin resistance, decrease in hyperglycemia, loss of body weight, reduction of blood pressure, decrease in reactive oxygen species (ROS) production, modulation of the inflammatory response, and improvement of reproductive function (Table 2, Figure 3) [38,44,45].
An endogenous GLP-1 is produced not only peripherally in the ileum, but also centrally in the brain, particularly in the area of nucleus of the solitary tract (NTS) of the hindbrain and the olfactory bulb, thereby controlling appetite and feeding behaviors [46]. The GLP-1 analog promotes satiety by activating GLP-1Rs in the hypothalamus and brainstem, resulting in a decline of food intake and weight loss [11,47]. Besides its metabolic functions in the brain, GLP-1 directly exhibits neuroprotective and neurotropic effects [48]. GLP-1 or exendin-4 administration into the lateral ventricles of mice reduced endogenous Aβ levels and prevented Aβ-induced cell death in rat hippocampus neurons [49]. During hypoxic injury, exendin-4 increased cell viability in cultured embryonic primary cerebral cortical and ventral mesencephalic (dopaminergic) neurons and protected against metabolic and oxidative damages. Importantly, these protective effects were not observed in neurons from GLP-1R knockout mice [50]. Exendin-4 also preserved cell viability and decreased apoptosis induced by hydrogen peroxide (H2O2)-induced oxidative stress in NSC19 neuroblastoma cells and in SOD1-G93A mutant mice models of amyotrophic lateral sclerosis [51,52].
The role of GLP-1 in the CNS, where it regulates satiety and body weight, is one of the intriguing aspects of its biology [47,53]. After activation of GLP-1Rs in the portal vein, this brain–gut axis regulates muscle glycogen synthesis and whole-body glucose clearance [54,55]. However, the relative importance of GLP-1Rs in the CNS and PNS in maintaining energy homeostasis is unknown. Previous studies have suggested that there is improved vascular function upon administration of GLP-1 in subjects with normal glucose tolerance and T2DM patients [56,57,58]. There was increased blood pressure and heart rate upon intermittent GLP-1 administration, while a modest increase in heart rate and a decrease in blood pressure was observed upon chronic activation of the GLP-1R with GLP-1 analogs [59]. This observation has raised the curiosity of many interested in the potential beneficial effects of GLP-1 on the heart. Evidently, GLP-1 and GLP-1Rs are abundantly distributed in many vital tissues, and their physiologic actions have gradually been elucidated, particularly since the advent of GLP-1 mimetics, including GLP-1R agonists and DPP-4 inhibitors.
GLP-1R expresses in various immune cells, including splenocytes, thymocytes, regulatory T cells, bone marrow-derived cells, macrophages, and invariant natural killer T cells, and thereby modulates the immune responses [60,61]. The GLP-1 analog, liraglutide, improved the psoriasis area and severity index and reduced the inflammatory cytokine secretion from natural killer T cells in patients with psoriasis [62]. In a mice model of obesity (high fat diet, HFD), liraglutide decreased cardiovascular risk factors including insulin resistance and inflammation, reduced monocyte vascular adhesion, and improved cardiac function by activating adenosine 5′-monophosphate activated protein kinase (AMPK) [63]. Moreover, liraglutide has been approved for chronic weight management in patients with obesity or weight related comorbid conditions including hyperlipidemia, hypertension, or T2DM [64,65]. Another GLP-1 analog, exndin-4, reduced mRNA levels of the proinflammatory cytokines, tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein 1 (MCP-1), and signal transducer and activator of transcription 3 (STAT-3) in HFD fed mice [66]. Additionally, the number of transforming growth factor-β (TGF- β) positive cells were increased upon GLP-1/gastrin treatment in islet transplanted nonobese diabetic animals [67].
GLP-1 analogs stimulate natriuresis at physiological levels by inhibiting sodium reabsorption via decreased activity of sodium–hydrogen exchanger 3 (NHE3) or solute carrier family 9 member 3 (SLC9A3) in the proximal tubules, resulting in decreased blood pressure in patients with diabetes [68,69]. It is yet to be confirmed whether this effect is caused by direct GLP-1R activation in the kidneys. However, the involvement of renin–angiotensin system and a neural pathway has been suggested by previous studies [68,69].

3. Molecular Mechanisms Underlying Cardioprotective Effects of GLP-1 and Its Analogs

Cardiac muscle is predisposed to damage and dysfunction under several metabolic stresses, particularly under diabetic conditions and biological aging. Based on intense lines of preclinical studies both in vitro (Table 3) and in vivo (Table 4), GLP-1 analogs expressed their cardiovascular benefits via several cytoprotective molecular mechanisms.

3.1. GLP-1 and Its Analogs and Cardiac Functions

Clinical research has demonstrated that GLP-1R analogs improve the prognosis of cardiovascular disorders by raising the resting heart rate and decreasing arrhythmia in patients with heart failure (HF) [25,90]. Patients with HF who received GLP-1R analog for 5 weeks have shown improvements in their left ventricular ejection fraction and 6-min walking distance as well as increases in their maximal oxygen uptake [91]. Albiglutide administration for 12 weeks in patients with HF elicits an improvement in their cardiac oxygen consumption [92]. Exenatide lowered atherosclerotic levels and enhanced diastolic function in patients with T2DM [93]. Mechanistically, GLP-1 analogs improve the sympathetic nervous system, which, in turn, causes an increase in heart rate [94,95]. While the systemic effects and increased heart rate elicited by exendin-4 treatment in vivo were eliminated by blocking β-adrenergic receptor (β-AR) [96], GLP-1 treatment via peripheral or intracerebroventricular routes dose-dependently increased blood pressure and heart rate in animal models [95,97,98]. The cause of an elevated heart rate has remained intriguing; however, it might be related to the stimulation of sympathetic nervous system. GLP-1 analogs (such as exendin-4) activated c-fos expression in the adrenal medulla and medullary catecholamine neurons, indicating that central GLP-1Rs mediate neuroendocrine responses and influence sympathetic outflow [95].
Cardiovascular risk factors such as dyslipidemia and hypertension have been demonstrated to be reduced by liraglutide and exenatide. Exenatide revealed a drop in systolic blood pressure (SBP) without changes in diastolic blood pressure (DBP) in a meta-analysis of six trials including over 2000 participants, while weight loss could not entirely explain this effect [99]. However, another clinical study demonstrated that liraglutide expresses a decrease in SBP in the first two weeks of treatment independent of weight loss [100]. Exenatide has been shown to lower triglyceride levels by 12%, total cholesterol by 5%, low-density lipoprotein cholesterol by 6%, and increase high-density lipoprotein cholesterol by 24% in three-year trials, thus contributing to lowering blood pressure [101].

3.2. GLP-1 and Its Analogs and Myocardial Glucose Uptake

GLP-1 has been suggested to have a wide range of biological effects on several target organs, including heart, liver, brain, muscles, and adipose tissues [44,102,103]. The classic response elicited by GLP-1R activation is the improvement of cardiac function by increasing glucose uptake and coronary flow as well as the secretion of the main blood pressure and electrolyte regulator, atrial natriuretic peptide (ANP), in mice [69,104,105]. GLP-1-(7-36) amide was found to directly augment myocardial glucose uptake by increasing the translocation of glucose transporters (GLUTs), GLUT-1 and GLUT-4, in normal and ischemia/reperfusion (I/R)-induced ischemic rat hearts in a similar pattern as insulin [106]. Treatment with exendin-4 induced cardiac glucose uptake and ATP production via an increase in GLUT-1 translocation which was dependent on the expression of p38 mitogen-activated protein kinase (p38MAPK) in hypoxia/reoxygenation (H/R)-induced injury in H9c2 cardiomyoblasts [107]. However, clinical evidence suggests that baseline functions of glucose transport and insulin resistance are the main determinants of the impact of GLP-1 analog on glucose transport and glucose uptake in cardiomyocytes during hyperglycemic states in patients with T2DM [108] and during normo- or hypoglycemia in healthy subjects [109]. The occurrence of apoptosis in cardiac cells contributes to the development of diabetic cardiomyopathy, which is caused by the buildup of intracellular lipids such as saturated palmitic acids. In addition, treatment with GLP-1 analogs provoked β-catenin, signaling leading to an activation of the protein kinase B-glycogen synthase kinase-3β survival pathway and a decrease in apoptotic activation in neonatal rat cardiomyocytes exposed to palmitate [110].

3.3. GLP-1 Analogs and Cardiac Oxidative Stress as Well as Ischemia/Reperfusion (I/R) Injury

Previous studies have shown that GLP-1 and its analogs express cardiac-protective effects and are crucial in the regulation of heart functions. Exendin-4, for instance, reduced infarct size and enhanced mechanical function in isolated rat hearts exposed to I/R injury [111]. Likewise, exenatide infusion decreased the size of the infarct area found in a porcine model of I/R injury [112]. In addition, ejection fraction and other functional indicators were improved in patients with acute myocardial infarction (MI) after a 72-h GLP-1 infusion [113]. Moreover, treatment with GLP-1 analogs prevented the cardiomyocyte damage and apoptosis from H/R injury [114]. Liraglutide inhibited plasminogen activator inhibitor type-1 (PAI-1) and vascular adhesion molecules in vitro and in vivo and enhanced NOS activity [1]. Moreover, GLP-1R activation restored endoplasmic reticulum (ER) homoeostasis, cytoprotection, and the signaling pathways disrupted by various stress stimuli [63,115,116].
Oxidative stress ensuing from an imbalance between antioxidant defenses and the generation of ROS could result in cellular damage and apoptosis. The toxic levels of ROS are generated in patients of HF [117] and animal models of MI [118]. Recent studies have shown that GLP-1 analogs exert cardioprotective benefits through regulating the antioxidant and anti-apoptotic responses. Activation of the GLP-1R increased cAMP levels, thereby activating PKA and Epac-dependent antioxidant and anti-apoptotic cytoprotective responses against H2O2-induced oxidative damage in cardiomyocytes [71]. Although it has been reported that excessive stimulation of β-ARs results in the induction of insulin resistance in many cell types, including cardiomyocytes [119,120], adipocytes [121], and skeletal muscle [122], prolonged overstimulation of GLP-1R has not been shown to cause these detrimental effects.
GLP-1 appears to have both GLP-1R-dependent and -independent mechanisms for mediating its cardioprotective and vasodilatory effects [123]. The findings that GLP-1(9-36) maintained its vasodilatory and cardioprotective properties in hearts isolated from GLP-1R−/− mice serve as an explanation of this phenomenon [123]. In fact, GLP-1(9-36) enhanced left ventricular function and boosted myocardial glucose absorption in dogs with dilated cardiomyopathy [16]. In contrast to intact GLP-1, GLP-1(9-36) directly inhibited the generation of superoxide in human artery endothelial cells induced by high glucose or free fatty acids [124] and cytoprotective actions on mouse cardiomyocytes exposed to H2O2 [115]. This is consistent with the findings that GLP-1(9-36) leads to significant enhancement of the rate-pressure product when the GLP-1R antagonist, exendin (9-39), is administered, although the GLP-1R agonist exendin-4, which is DPP4 resistant, is no longer effective in increasing left ventricular end-diastolic pressure in isolated rat hearts [111], indicating the presence of a GLP-1R independent pathway. This is further supported by our recent findings, wherein exposure to GLP-1(9-36) inhibited H2O2-induced oxidative stress and apoptosis in H9c2 cardiomyoblasts via PI3K/Akt/NOS signaling pathway; therefore, GLP-1R analog treatment represents a potential therapeutic approach for the prevention of pathological oxidative stress and damage to the heart [15].

4. Significant Roles of GLP-1 and Its Analogs in Mitochondrial Homeostasis

Mitochondria is the energy generator for the cell, oxidizing various carbon sources and producing energy in the form of ATP. Mitochondria produces intermediate substrates to be utilized in anabolic or catabolic processes and regulates crucial physiological and pathological processes such as calcium homeostasis, metabolism of biomolecules (amino acids, lipids, and glucose), oxidative stress, apoptosis, aging, and autophagy [125]. The majority of the energy for cardiac functions is derived from the oxidation of fatty acids in mitochondria. However, in T2DM, insulin resistance leads to lower glucose utilization and oxidative phosphorylation; consequently, cardiomyocytes become reliant on the oxidation of fatty acids for energy supply. This shift in substrate utilization leads to an imbalance in the uptake and oxidation of fatty acids, which ultimately causes mitochondrial dysfunction. Moreover, chronic hyperglycemia instigates mitochondrial oxidative stress and fragmentation, leading to cellular dysfunction and apoptosis [2]. In diabetic hearts, the damaged mitochondria in cardiomyocytes resulted in the generation of excessive amounts of ROS causing pathological oxidative stress, which deteriorated the myocardial functions [126].
Therefore, repair of the damaged mitochondria and replenishment of new mitochondria would be an effective strategy to maintain a healthy pool of mitochondria to ameliorate the pathological outcomes of cardiac diseases. Our recent study showed that stimulation of GLP-1R using exendin-4 exhibits antioxidant and antiapoptotic effects, as well as an improvement in mitochondrial functions via cAMP/Epac/PI3K/Akt signaling in H9c2 cardiomyoblasts exposed with methylglyoxal, an advanced glycation end product (AGE). Since intracellularly accumulation of AGEs causes mitochondrial impairment, oxidative stress, and apoptosis, attenuating methylglyoxal-induced mitochondrial abnormalities in the heart by exendin-4 may help to ameliorate and delay the progression of heart diseases [70]. Several clinical studies have also supported that GLP-1 analogs are beneficial in lowering the occurrence of adverse cardiovascular events [127,128]. GLP-1 analogs with longer half-life, such as exenatide and liraglutide, have been designed to tolerate DPP-4 degradation and are efficacious in treating T2DM [129,130]. However, long-term usage is linked to an increased risk of hypoglycemia, pancreatic cancer, thyroid cancer, and gastric ailments [131,132].
Although the precise molecular mechanisms responsible for the cardioprotective benefits of GLP-1 analogs have not been clearly identified so far, there are preclinical studies (Table 3 and Table 4) which suggest that this drug class may also be effective when administered shortly after MI through the activation of the mitophagy process and the resurrection of non-performing mitochondria. In H/R model of H9c2 cells, pretreatment with exenatide reduced mitochondrial abnormalities by diminishing oxidative stress and mitochondrial calcium overload while increasing mitochondrial ATP synthase activity or ATP synthesis and rescuing mitochondrial membrane potential (MMP) [133]. Moreover, mitochondrial uncoupling protein-3 (UCP-3) and nuclear respiratory factor-1 (NRF-1) were increased while lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), and cardiomyocyte apoptosis were decreased [133]. Liraglutide has been reported to protect cardiomyocytes from interleukin-1β (IL-1β)-induced metabolic disturbance and mitochondrial dysfunction by enhancing phosphorylated AMPK and acetyl-coA carboxylase (ACC) as well as improving peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), carnitine palmitoyltransferase-1 (CPT1), and diacylglycerol acyltransferase 1 (DGAT1) [134]. GLP-1 analog also decreased the expression of H/R-induced ER stress proteins, including glucose regulatory protein 78 (GRP78), CCAAT-enhancer-binding protein homologous protein (CHOP), and caspase-12, and exhibited cardioprotective effects by an activation of the GLP-1R/PI3K/Akt signaling pathway [114].
Hyperglycemic condition in primary cardiomyocytes has been shown to cause ER stress, mitochondrial oxidative stress, and enhanced mitochondria-associated ER membrane (MAMs)-associated proteins, including GRP75 and mitofusin-2 (MFN2). MFN2 played an important role in high glucose-induced ER stress in cardiomyocytes, while silencing of MFN2 abated mitochondrial calcium overload-mediated mitochondrial impairment, thereby decreasing ER stress-mediated cardiomyocyte apoptosis [135]. Furthermore, metabolic signaling pathways mediated by insulin-like growth factor 1 (IGF-1), mammalian target of rapamycin (mTOR), AMPK, and sirtuins are linked to the balance between nutrient anabolism and catabolism in cells, which maintain cellular homeostasis [136,137]. In both in vivo and in vitro studies, employing cardiac hypertensive model elucidated that administration of exendin-4 attenuated cardiac hypertrophy via the inhibition of mitochondrial permeability transition pore (MPTP) opening and ROS release [138].
Exendin-4 also was found to inhibit cardiac hypertrophy by upregulating GLP-1R expression and activating the AMPK signaling pathway [72]. Liraglutide could protect the heart against aortic banding-induced myocardial fibrosis and dysfunction by suppressing mTOR/p70S6K signaling and boosting autophagy [89]. Another study reported that liraglutide reduces cardiac hypertrophy via the angiotensin II/AT1R/ACE2 and AMPK/mTOR/ p70S6K pathways [76]. A recent study examined the efficacy of a newly synthesized GLP-1R agonist, compound DMB, in attenuating pathological left ventricular remodeling in a permanent coronary artery ligation mice model and suggested that increasing mitochondrial biogenesis and Parkin-associated mitophagy after MI might be a viable target for the therapeutic management of adverse cardiac remodeling [139]. In line with previous studies, liraglutide ameliorated cardiac fibrosis and cardiomyocyte death in the infarcted heart in rat, while their mechanistic findings in H9c2 cardiomyoblasts showed that SIRT1/Parkin/mitophagy pathways were involved in its cytoprotective effects [140]. Contrastingly, another study did not find any effects on adverse remodeling or cardiac function upon a similar liraglutide treatment in rats [140]. Nevertheless, excessive glucose exposure caused increased intracellular ROS, apoptosis, MMP loss, decreased autophagy, and increased phospho-mTOR (p-mTOR) and p-ULK1 levels, all of which were counteracted by GLP-1 analogs. These findings suggested that GLP-1 analogs could reverse glucose toxicity by inducing mTOR/ULK1-dependent autophagy [141].

5. Co-Agonists: A Single Molecule Stimulating Multiple Peptide Receptors

There have been propositions that multiple signaling pathways would need to be targeted for an efficient clinical treatment of obesity due to the system’s redundancy in regulating food intake and body weight [142]. Using this concept, the most delicate way to treat T2DM was to combine activity of GLP-1 with that of the GIP to achieve long-lasting beneficial effects. A synthetic peptide with stimulatory activity at both the GIP receptor and GLP-1R proved to be more effective at reducing body weight and blood glucose in obese mice compared to a selective GLP-1R agonist [143]. Additionally, recent studies in mice have demonstrated that GIP receptor is expressed in hypothalamic neurons, and that activating it significantly reduced food intake and body weight, when combined with GLP-1 [144,145,146]. These initial observations, together with subsequent studies, led to the development of multi-receptor peptides affecting more than one intracellular signaling pathway, which have become the choice of treatments for T2DM and obesity.
Following the initial studies in multi-receptor peptides, a number of single-molecule GIP receptors and GLP-1R agonists were created. Tirzepatide, a dual GIP/GLP-1 receptor co-agonist for the treatment of T2DM, is a 39-amino acid linear peptide that is comparable in size to the related hormones GIP and GLP-1 [147]. In murine pancreatic islets from GIP receptor knockout mice, GLP-1R antagonist inhibited the insulin secretion stimulated by tirzepatide, while in islets lacking GLP-1Rs, GIP receptor blocker suppressed the insulin secretion upon tirzepatide stimulation. Moreover, tirzepatide decreased both food intake and body weight more prominently than selective GLP-1R agonists such as semaglutide in HFD mice [51]. Tirzepatide stimulated cAMP production in pancreatic ß-cells, similar to a combination of GLP-1 and GIP [51], while also eliciting better efficacy in controlling cardiovascular disease risk factors such as glucose levels and body weight loss in diabetic patients [148,149].
Tirzepatide ameliorated a number of cardiovascular risk factors over selective GLP-1R agonists. In blood samples analyzed from diabetic patients, triglycerides, apolipoprotein C-III, phosphatidylethanolamines/phosphatidylcholines, circulating levels of branched-chain amino acids (leucine, isoleucine, and valine), inflammatory markers (e.g., C-reactive protein), and adhesion molecules (e.g., intercellular adhesion molecule-1; ICAM-1) were reduced by tirzepatide [150,151,152]. Additionally, a preliminary proof of tirzepatide’s cardiovascular safety was elucidated by a recent meta-analysis combining all the clinical trials. The commonly specified cardiovascular end points were projected to have a risk below 1.0, and for 4-point major adverse cardiovascular events (MACE) (MI, stroke, hospitalization for angina, and all-cause death) and 3-point MACE (MI, stroke, and all-cause mortality), the hazard ratios were less than 1.3 [153].

6. Conclusions and Future Directions

Inadequate exercise, high fat and highly processed diet, and excess nutrition all contribute to the rising prevalence of T2DM, which severely reduces both life expectancy and quality of life. T2DM is linked to a large worldwide mortality burden, which primarily involves cardiovascular events including HF and stroke. Therefore, new therapeutic modalities are highly required. GLP-1 analogs are relatively novel medications used for the treatment of T2DM. We summarized the recent findings pertaining to cardioprotective effects as well as molecular mechanisms of GLP-1R-dependent and -independent pathways (Figure 4). GLP-1 analogs are able to directly affect not only other important cardiovascular risk factors, including high blood pressure, dyslipidemia, or obesity, but also the hyperglycemia in diabetic patients. These characteristics were exemplified in several cardiovascular outcome trials for exendin-4, liraglutide, semaglutide, dulaglutide, albiglutide, and tirzepatide, all of which demonstrated the improvement of cardiovascular prognosis in addition to safety. Although the effects of the GLP-1R agonists were well understood, further research is necessary to fully understand their GLP-1R-dependent and -independent effects on the cardiovascular system, particularly with regard to their signaling pathways. Recently, GLP-1R-independent pathways have been identified to have direct vasodilatory and cardioprotective effects, as well as anti-inflammatory and antioxidant benefits. The protective properties of GLP-1 analogs can be thoroughly investigated using contemporary omics technologies to reveal epigenetic, metabolomics, and proteomic changes to explain the observed beneficial effects. The discovery of additional GLP-1 signaling pathways (unrelated to glucose lowering effects) is very likely to make this class of medication relevant for the treatment of several chronic diseases, including obesity, T2DM, cardiovascular diseases, cancer, neurodegenerative and neuroinflammatory diseases, and others.

Author Contributions

Conceptualization, S.P., S.M. and W.P.; writing—original draft preparation, S.P.; writing—review and editing, S.M. and W.P.; visualization, S.P.; supervision, W.P.; funding acquisition, W.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research project was supported by grants from the National Research Council of Thailand (NRCT) and Mahidol University (Grant number: N42A650352) (to W.P.) and the Specific League Funds from Mahidol University (Grant number: SLF-2 MUPY 01/2564) (to W.P). S.P. is currently funded by an International Postdoctoral Fellowship from Mahidol University.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analyzed during the current study are included in this published article.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Del Olmo-Garcia, M.I.; Merino-Torres, J.F. GLP-1 receptor agonists and cardiovascular disease in patients with type 2 diabetes. J. Diabetes Res. 2018, 2018, 4020492. [Google Scholar] [CrossRef] [Green Version]
  2. Jia, G.; Hill, M.A.; Sowers, J.R. Diabetic cardiomyopathy: An update of mechanisms contributing to this clinical entity. Circ. Res. 2018, 122, 624–638. [Google Scholar] [CrossRef]
  3. Campbell, J.E.; Drucker, D.J. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013, 17, 819–837. [Google Scholar] [CrossRef] [Green Version]
  4. Graaf, C.; Donnelly, D.; Wootten, D.; Lau, J.; Sexton, P.M.; Miller, L.J.; Ahn, J.M.; Liao, J.; Fletcher, M.M.; Yang, D.; et al. Glucagon-like peptide-1 and its class B G protein-coupled receptors: A long march to therapeutic successes. Pharmacol. Rev. 2016, 68, 954–1013. [Google Scholar] [CrossRef] [Green Version]
  5. Moore, B. On the treatment of diabetus mellitus by acid extract of duodenal mucous membrane. Biochem. J. 1906, 1, 28–38. [Google Scholar] [CrossRef] [Green Version]
  6. Brown, J.C.; Dryburgh, J.R.; Ross, S.A.; Dupre, J. Identification and actions of gastric inhibitory polypeptide. Recent Prog. Horm. Res. 1975, 31, 487–532. [Google Scholar] [CrossRef]
  7. Sarson, D.L.; Wood, S.M.; Kansal, P.C.; Bloom, S.R. Glucose-dependent insulinotropic polypeptide augmentation of insulin. Physiology or pharmacology? Diabetes 1984, 33, 389–393. [Google Scholar] [CrossRef]
  8. Bell, G.I.; Santerre, R.F.; Mullenbach, G.T. Hamster preproglucagon contains the sequence of glucagon and two related peptides. Nature 1983, 302, 716–718. [Google Scholar] [CrossRef]
  9. Shields, D.; Warren, T.G.; Roth, S.E.; Brenner, M.J. Cell-free synthesis and processing of multiple precursors to glucagon. Nature 1981, 289, 511–514. [Google Scholar] [CrossRef]
  10. Orskov, C.; Bersani, M.; Johnsen, A.H.; Hojrup, P.; Holst, J.J. Complete sequences of glucagon-like peptide-1 from human and pig small intestine. J. Biol. Chem. 1989, 264, 12826–12829. [Google Scholar] [CrossRef]
  11. Sandoval, D.A.; D’Alessio, D.A. Physiology of proglucagon peptides: Role of glucagon and GLP-1 in health and disease. Physiol. Rev. 2015, 95, 513–548. [Google Scholar] [CrossRef] [Green Version]
  12. Doyle, M.E.; Egan, J.M. Mechanisms of action of glucagon-like peptide 1 in the pancreas. Pharmacol. Ther. 2007, 113, 546–593. [Google Scholar] [CrossRef] [Green Version]
  13. Meloni, A.R.; DeYoung, M.B.; Lowe, C.; Parkes, D.G. GLP-1 receptor activated insulin secretion from pancreatic beta-cells: Mechanism and glucose dependence. Diabetes Obes. Metab. 2013, 15, 15–27. [Google Scholar] [CrossRef] [Green Version]
  14. Rolin, B.; Deacon, C.F.; Carr, R.D.; Ahren, B. The major glucagon-like peptide-1 metabolite, GLP-1-(9-36)-amide, does not affect glucose or insulin levels in mice. Eur. J. Pharmacol. 2004, 494, 283–288. [Google Scholar] [CrossRef]
  15. Nuamnaichati, N.; Parichatikanond, W.; Mangmool, S. Cardioprotective effects of glucagon-like peptide-1 (9-36) against oxidative injury in H9c2 cardiomyoblasts: Potential role of the PI3K/Akt/NOS pathway. J. Cardiovasc. Pharmacol. 2022, 79, e50–e63. [Google Scholar] [CrossRef]
  16. Nikolaidis, L.A.; Elahi, D.; Shen, Y.T.; Shannon, R.P. Active metabolite of GLP-1 mediates myocardial glucose uptake and improves left ventricular performance in conscious dogs with dilated cardiomyopathy. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, H2401–H2408. [Google Scholar] [CrossRef] [Green Version]
  17. Picatoste, B.; Ramirez, E.; Caro-Vadillo, A.; Iborra, C.; Ares-Carrasco, S.; Egido, J.; Tunon, J.; Lorenzo, O. Sitagliptin reduces cardiac apoptosis, hypertrophy and fibrosis primarily by insulin-dependent mechanisms in experimental type-II diabetes. Potential roles of GLP-1 isoforms. PLoS ONE 2013, 8, e78330. [Google Scholar] [CrossRef]
  18. Poudyal, H. Mechanisms for the cardiovascular effects of glucagon-like peptide-1. Acta Physiol. 2016, 216, 277–313. [Google Scholar] [CrossRef]
  19. Deacon, C.F.; Johnsen, A.H.; Holst, J.J. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J. Clin. Endocrinol. Metab. 1995, 80, 952–957. [Google Scholar] [CrossRef]
  20. Pabreja, K.; Mohd, M.A.; Koole, C.; Wootten, D.; Furness, S.G. Molecular mechanisms underlying physiological and receptor pleiotropic effects mediated by GLP-1R activation. Br. J. Pharmacol. 2014, 171, 1114–1128. [Google Scholar] [CrossRef] [Green Version]
  21. Tomas, E.; Habener, J.F. Insulin-like actions of glucagon-like peptide-1: A dual receptor hypothesis. Trends Endocrinol. Metab. 2010, 21, 59–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Liu, Z.; Stanojevic, V.; Brindamour, L.J.; Habener, J.F. GLP1-derived nonapeptide GLP1(28-36)amide protects pancreatic beta-cells from glucolipotoxicity. J. Endocrinol. 2012, 213, 143–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tomas, E.; Stanojevic, V.; Habener, J.F. GLP-1-derived nonapeptide GLP-1(28-36)amide targets to mitochondria and suppresses glucose production and oxidative stress in isolated mouse hepatocytes. Regul. Pept. 2011, 167, 177–184. [Google Scholar] [CrossRef] [PubMed]
  24. Nauck, M.A.; Vardarli, I.; Deacon, C.F.; Holst, J.J.; Meier, J.J. Secretion of glucagon-like peptide-1 (GLP-1) in type 2 diabetes: What is up, what is down? Diabetologia 2011, 54, 10–18. [Google Scholar] [CrossRef] [Green Version]
  25. Sonne, D.P.; Rehfeld, J.F.; Holst, J.J.; Vilsboll, T.; Knop, F.K. Postprandial gallbladder emptying in patients with type 2 diabetes: Potential implications for bile-induced secretion of glucagon-like peptide 1. Eur. J. Endocrinol. 2014, 171, 407–419. [Google Scholar] [CrossRef] [Green Version]
  26. Guglielmi, V.; Sbraccia, P. GLP-1 receptor independent pathways: Emerging beneficial effects of GLP-1 breakdown products. Eat. Weight Disord. 2017, 22, 231–240. [Google Scholar] [CrossRef]
  27. Gupta, V. Glucagon-like peptide-1 analogues: An overview. Indian J. Endocrinol. Metab. 2013, 17, 413–421. [Google Scholar] [CrossRef]
  28. Kalra, S.; Baruah, M.P.; Sahay, R.K.; Unnikrishnan, A.G.; Uppal, S.; Adetunji, O. Glucagon-like peptide-1 receptor agonists in the treatment of type 2 diabetes: Past, present, and future. Indian J. Endocrinol. Metab. 2016, 20, 254–267. [Google Scholar] [CrossRef]
  29. Gilbert, M.P.; Pratley, R.E. GLP-1 analogs and DPP-4 inhibitors in type 2 diabetes therapy: Review of head-to-head clinical trials. Front. Endocrinol. 2020, 11, 178. [Google Scholar] [CrossRef]
  30. Htike, Z.Z.; Zaccardi, F.; Papamargaritis, D.; Webb, D.R.; Khunti, K.; Davies, M.J. Efficacy and safety of glucagon-like peptide-1 receptor agonists in type 2 diabetes: A systematic review and mixed-treatment comparison analysis. Diabetes Obes. Metab. 2017, 19, 524–536. [Google Scholar] [CrossRef] [Green Version]
  31. MacDonald, P.E.; El-Kholy, W.; Riedel, M.J.; Salapatek, A.M.; Light, P.E.; Wheeler, M.B. The multiple actions of GLP-1 on the process of glucose-stimulated insulin secretion. Diabetes 2002, 51 (Suppl. S3), S434–S442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Park, S.; Dong, X.; Fisher, T.L.; Dunn, S.; Omer, A.K.; Weir, G.; White, M.F. Exendin-4 uses Irs2 signaling to mediate pancreatic beta cell growth and function. J. Biol. Chem. 2006, 281, 1159–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Portha, B.; Tourrel-Cuzin, C.; Movassat, J. Activation of the GLP-1 receptor signalling pathway: A relevant strategy to repair a deficient beta-cell mass. Exp. Diabetes Res. 2011, 2011, 376509. [Google Scholar] [CrossRef] [Green Version]
  34. Meurot, C.; Jacques, C.; Martin, C.; Sudre, L.; Breton, J.; Rattenbach, R.; Bismuth, K.; Berenbaum, F. Targeting the GLP-1/GLP-1R axis to treat osteoarthritis: A new opportunity? J. Orthop. Translat. 2022, 32, 121–129. [Google Scholar] [CrossRef] [PubMed]
  35. Reed, J.; Bain, S.; Kanamarlapudi, V. Recent advances in understanding the role of glucagon-like peptide 1. F1000Res 2020, 9, 239. [Google Scholar] [CrossRef] [Green Version]
  36. Peng, W.; Zhou, R.; Sun, Z.F.; Long, J.W.; Gong, Y.Q. Novel insights into the roles and mechanisms of GLP-1 receptor agonists against aging-related diseases. Aging Dis. 2022, 13, 468–490. [Google Scholar] [CrossRef]
  37. Pratley, R.E.; Jacob, S.; Baek, S.; Trautmann, M.E.; Hompesch, M.; Han, O.; Stewart, J.; Sorli, C.H.; Shaunik, A.; Yoon, K.H. Efficacy and safety of efpeglenatide in key patient subgroups from the BALANCE randomized trial, stratified by pre-diabetes status, BMI, and age at baseline. BMJ Open Diabetes Res. Care 2022, 10, e002207. [Google Scholar] [CrossRef]
  38. Andersen, A.; Lund, A.; Knop, F.K.; Vilsboll, T. Glucagon-like peptide 1 in health and disease. Nat. Rev. Endocrinol. 2018, 14, 390–403. [Google Scholar] [CrossRef]
  39. Sposito, A.C.; Berwanger, O.; de Carvalho, L.S.F.; Saraiva, J.F.K. GLP-1RAs in type 2 diabetes: Mechanisms that underlie cardiovascular effects and overview of cardiovascular outcome data. Cardiovasc. Diabetol. 2018, 17, 157. [Google Scholar] [CrossRef]
  40. Wettergren, A.; Schjoldager, B.; Mortensen, P.E.; Myhre, J.; Christiansen, J.; Holst, J.J. Truncated GLP-1 (proglucagon 78-107-amide) inhibits gastric and pancreatic functions in man. Dig. Dis. Sci. 1993, 38, 665–673. [Google Scholar] [CrossRef]
  41. Maljaars, P.W.; Peters, H.P.; Mela, D.J.; Masclee, A.A. Ileal brake: A sensible food target for appetite control. A review. Physiol. Behav. 2008, 95, 271–281. [Google Scholar] [CrossRef] [PubMed]
  42. Schirra, J.; Nicolaus, M.; Roggel, R.; Katschinski, M.; Storr, M.; Woerle, H.J.; Goke, B. Endogenous glucagon-like peptide 1 controls endocrine pancreatic secretion and antro-pyloro-duodenal motility in humans. Gut 2006, 55, 243–251. [Google Scholar] [CrossRef] [Green Version]
  43. Wettergren, A.; Petersen, H.; Orskov, C.; Christiansen, J.; Sheikh, S.P.; Holst, J.J. Glucagon-like peptide-1 7-36 amide and peptide YY from the L-cell of the ileal mucosa are potent inhibitors of vagally induced gastric acid secretion in man. Scand. J. Gastroenterol. 1994, 29, 501–505. [Google Scholar] [CrossRef]
  44. Rowlands, J.; Heng, J.; Newsholme, P.; Carlessi, R. Pleiotropic effects of GLP-1 and analogs on cell signaling, metabolism, and function. Front. Endocrinol. 2018, 9, 672. [Google Scholar] [CrossRef] [Green Version]
  45. Papaetis, G.S.; Kyriacou, A. GLP-1 receptor agonists, polycystic ovary syndrome and reproductive dysfunction: Current research and future horizons. Adv. Clin. Exp. Med. 2022, 31, 1265–1274. [Google Scholar] [CrossRef] [PubMed]
  46. Daniels, D.; Mietlicki-Baase, E.G. Glucagon-Like Peptide 1 in the Brain: Where Is It Coming From, Where Is It Going? Diabetes 2019, 68, 15–17. [Google Scholar] [CrossRef] [Green Version]
  47. Holst, J.J. The physiology of glucagon-like peptide 1. Physiol. Rev. 2007, 87, 1409–1439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Heppner, K.M.; Perez-Tilve, D. GLP-1 based therapeutics: Simultaneously combating T2DM and obesity. Front. Neurosci. 2015, 9, 92. [Google Scholar] [CrossRef] [Green Version]
  49. Perry, T.; Lahiri, D.K.; Sambamurti, K.; Chen, D.; Mattson, M.P.; Egan, J.M.; Greig, N.H. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. J. Neurosci. Res. 2003, 72, 603–612. [Google Scholar] [CrossRef]
  50. Li, Y.; Perry, T.; Kindy, M.S.; Harvey, B.K.; Tweedie, D.; Holloway, H.W.; Powers, K.; Shen, H.; Egan, J.M.; Sambamurti, K.; et al. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc. Natl. Acad. Sci. USA 2009, 106, 1285–1290. [Google Scholar] [CrossRef] [Green Version]
  51. Coskun, T.; Sloop, K.W.; Loghin, C.; Alsina-Fernandez, J.; Urva, S.; Bokvist, K.B.; Cui, X.; Briere, D.A.; Cabrera, O.; Roell, W.C.; et al. LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: From discovery to clinical proof of concept. Mol. Metab. 2018, 18, 3–14. [Google Scholar] [CrossRef] [PubMed]
  52. Li, Y.; Chigurupati, S.; Holloway, H.W.; Mughal, M.; Tweedie, D.; Bruestle, D.A.; Mattson, M.P.; Wang, Y.; Harvey, B.K.; Ray, B.; et al. Exendin-4 ameliorates motor neuron degeneration in cellular and animal models of amyotrophic lateral sclerosis. PLoS ONE 2012, 7, e32008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Baggio, L.L.; Drucker, D.J. Glucagon-like peptide-1 receptors in the brain: Controlling food intake and body weight. J. Clin. Investig. 2014, 124, 4223–4226. [Google Scholar] [CrossRef] [Green Version]
  54. Burcelin, R.; Da Costa, A.; Drucker, D.; Thorens, B. Glucose competence of the hepatoportal vein sensor requires the presence of an activated glucagon-like peptide-1 receptor. Diabetes 2001, 50, 1720–1728. [Google Scholar] [CrossRef] [Green Version]
  55. Knauf, C.; Cani, P.D.; Kim, D.H.; Iglesias, M.A.; Chabo, C.; Waget, A.; Colom, A.; Rastrelli, S.; Delzenne, N.M.; Drucker, D.J.; et al. Role of central nervous system glucagon-like peptide-1 receptors in enteric glucose sensing. Diabetes 2008, 57, 2603–2612. [Google Scholar] [CrossRef] [Green Version]
  56. Almutairi, M.; Al Batran, R.; Ussher, J.R. Glucagon-like peptide-1 receptor action in the vasculature. Peptides 2019, 111, 26–32. [Google Scholar] [CrossRef]
  57. Forst, T.; Weber, M.M.; Pfutzner, A. Cardiovascular benefits of GLP-1-based herapies in patients with diabetes mellitus type 2: Effects on endothelial and vascular dysfunction beyond glycemic control. Exp. Diabetes Res. 2012, 2012, 635472. [Google Scholar] [CrossRef]
  58. Reed, J.; Bain, S.; Kanamarlapudi, V. A review of current trends with type 2 diabetes epidemiology, aetiology, pathogenesis, treatments and future perspectives. Diabetes Metab. Syndr. Obes. 2021, 14, 3567–3602. [Google Scholar] [CrossRef]
  59. Drucker, D.J. The cardiovascular biology of glucagon-like peptide-1. Cell Metab. 2016, 24, 15–30. [Google Scholar] [CrossRef]
  60. Hadjiyanni, I.; Siminovitch, K.A.; Danska, J.S.; Drucker, D.J. Glucagon-like peptide-1 receptor signalling selectively regulates murine lymphocyte proliferation and maintenance of peripheral regulatory T cells. Diabetologia 2010, 53, 730–740. [Google Scholar] [CrossRef] [Green Version]
  61. Panjwani, N.; Mulvihill, E.E.; Longuet, C.; Yusta, B.; Campbell, J.E.; Brown, T.J.; Streutker, C.; Holland, D.; Cao, X.; Baggio, L.L.; et al. GLP-1 receptor activation indirectly reduces hepatic lipid accumulation but does not attenuate development of atherosclerosis in diabetic male ApoE(-/-) mice. Endocrinology 2013, 154, 127–139. [Google Scholar] [CrossRef] [Green Version]
  62. Xu, X.; Lin, L.; Chen, P.; Yu, Y.; Chen, S.; Chen, X.; Shao, Z. Treatment with liraglutide, a glucagon-like peptide-1 analogue, improves effectively the skin lesions of psoriasis patients with type 2 diabetes: A prospective cohort study. Diabetes Res. Clin. Pract. 2019, 150, 167–173. [Google Scholar] [CrossRef] [PubMed]
  63. Noyan-Ashraf, M.H.; Shikatani, E.A.; Schuiki, I.; Mukovozov, I.; Wu, J.; Li, R.K.; Volchuk, A.; Robinson, L.A.; Billia, F.; Drucker, D.J.; et al. A glucagon-like peptide-1 analog reverses the molecular pathology and cardiac dysfunction of a mouse model of obesity. Circulation 2013, 127, 74–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mehta, A.; Marso, S.P.; Neeland, I.J. Liraglutide for weight management: A critical review of the evidence. Obes. Sci. Pract. 2017, 3, 3–14. [Google Scholar] [CrossRef]
  65. Whitten, J.S. Liraglutide (Saxenda) for weight loss. Am. Fam. Physician 2016, 94, 161–166. [Google Scholar]
  66. Koehler, J.A.; Baggio, L.L.; Lamont, B.J.; Ali, S.; Drucker, D.J. Glucagon-like peptide-1 receptor activation modulates pancreatitis-associated gene expression but does not modify the susceptibility to experimental pancreatitis in mice. Diabetes 2009, 58, 2148–2161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Suarez-Pinzon, W.L.; Power, R.F.; Yan, Y.; Wasserfall, C.; Atkinson, M.; Rabinovitch, A. Combination therapy with glucagon-like peptide-1 and gastrin restores normoglycemia in diabetic NOD mice. Diabetes 2008, 57, 3281–3288. [Google Scholar] [CrossRef]
  68. Muskiet, M.H.A.; Tonneijck, L.; Smits, M.M.; van Baar, M.J.B.; Kramer, M.H.H.; Hoorn, E.J.; Joles, J.A.; van Raalte, D.H. GLP-1 and the kidney: From physiology to pharmacology and outcomes in diabetes. Nat. Rev. Nephrol. 2017, 13, 605–628. [Google Scholar] [CrossRef]
  69. Skov, J. Effects of GLP-1 in the kidney. Rev. Endocr. Metab. Disord. 2014, 15, 197–207. [Google Scholar] [CrossRef]
  70. Nuamnaichati, N.; Mangmool, S.; Chattipakorn, N.; Parichatikanond, W. Stimulation of GLP-1 receptor inhibits methylglyoxal-induced mitochondrial dysfunctions in H9c2 cardiomyoblasts: Potential role of Epac/PI3K/Akt pathway. Front. Pharmacol. 2020, 11, 805. [Google Scholar] [CrossRef]
  71. Mangmool, S.; Hemplueksa, P.; Parichatikanond, W.; Chattipakorn, N. Epac is required for GLP-1R-mediated inhibition of oxidative stress and apoptosis in cardiomyocytes. Mol. Endocrinol. 2015, 29, 583–596. [Google Scholar] [CrossRef] [PubMed]
  72. Zhou, Y.; He, X.; Chen, Y.; Huang, Y.; Wu, L.; He, J. Exendin-4 attenuates cardiac hypertrophy via AMPK/mTOR signaling pathway activation. Biochem. Biophys. Res. Commun. 2015, 468, 394–399. [Google Scholar] [CrossRef]
  73. He, W.; Tong, G.; Fan, H.; Zhen, C.; Zeng, L.; Xue, L.; Chen, J.; Sun, Z.; He, P. Exendin-4 alleviates myocardial ischemia reperfusion injury by enhancing autophagy through promoting nuclear translocation of TFEB. Exp. Cell Res. 2023, 423, 113469. [Google Scholar] [CrossRef] [PubMed]
  74. Lyu, J.; Imachi, H.; Fukunaga, K.; Sato, S.; Kobayashi, T.; Saheki, T.; Japar, S.; Iwama, H.; Matsumura, Y.; Ozaki, M.; et al. Exendin-4 increases scavenger receptor class BI expression via activation of AMPK/FoxO1 in human vascular endothelial cells. Curr. Issues Mol. Biol. 2022, 44, 5474–5484. [Google Scholar] [CrossRef] [PubMed]
  75. Huang, J.H.; Chen, Y.C.; Lee, T.I.; Kao, Y.H.; Chazo, T.F.; Chen, S.A.; Chen, Y.J. Glucagon-like peptide-1 regulates calcium homeostasis and electrophysiological activities of HL-1 cardiomyocytes. Peptides 2016, 78, 91–98. [Google Scholar] [CrossRef]
  76. Wang, J.; Fan, S.; Xiong, Q.; Niu, Y.; Zhang, X.; Qin, J.; Shi, Y.; Zhang, L. Glucagon-like peptide-1 attenuates cardiac hypertrophy via the AngII/AT1R/ACE2 and AMPK/mTOR/p70S6K pathways. Acta Biochim. Biophys. Sin. 2021, 53, 1189–1197. [Google Scholar] [CrossRef]
  77. Li, Q.; Tuo, X.; Li, B.; Deng, Z.; Qiu, Y.; Xie, H. Semaglutide attenuates excessive exercise-induced myocardial injury through inhibiting oxidative stress and inflammation in rats. Life Sci. 2020, 250, 117531. [Google Scholar] [CrossRef]
  78. Zhu, Q.; Luo, Y.; Wen, Y.; Wang, D.; Li, J.; Fan, Z. Semaglutide inhibits ischemia/reperfusion-induced cardiomyocyte apoptosis through activating PKG/PKCepsilon/ERK1/2 pathway. Biochem. Biophys. Res. Commun. 2023, 647, 1–8. [Google Scholar] [CrossRef]
  79. Chen, J.; Wang, D.; Wang, F.; Shi, S.; Chen, Y.; Yang, B.; Tang, Y.; Huang, C. Exendin-4 inhibits structural remodeling and improves Ca2+ homeostasis in rats with heart failure via the GLP-1 receptor through the eNOS/cGMP/PKG pathway. Peptides 2017, 90, 69–77. [Google Scholar] [CrossRef]
  80. Chen, J.; Xu, S.; Wang, L.; Zhou, W.; Li, P.; Deng, N.; Tang, Q.; Li, Y.; Wu, L.; Chen, J.; et al. Exendin-4 inhibits atrial arrhythmogenesis in a model of myocardial infarction-induced heart failure via the GLP-1 receptor signaling pathway. Exp. Ther. Med. 2020, 20, 3669–3678. [Google Scholar] [CrossRef]
  81. Wei, H.; Bu, R.; Yang, Q.; Jia, J.; Li, T.; Wang, Q.; Chen, Y. Exendin-4 protects against hyperglycemia-induced cardiomyocyte pyroptosis via the AMPK-TXNIP pathway. J. Diabetes Res. 2019, 2019, 8905917. [Google Scholar] [CrossRef] [PubMed]
  82. Eid, R.A.; Khalil, M.A.; Alkhateeb, M.A.; Eleawa, S.M.; Zaki, M.S.A.; El-Kott, A.F.; Al-Shraim, M.; El-Sayed, F.; Eldeen, M.A.; Bin-Meferij, M.M.; et al. Exendin-4 attenuates remodeling in the remote myocardium of rats after an acute myocardial infarction by activating beta-arrestin-2, protein phosphatase 2A, and glycogen synthase kinase-3 and inhibiting beta-catenin. Cardiovasc. Drugs Ther. 2021, 35, 1095–1110. [Google Scholar] [CrossRef] [PubMed]
  83. Wassef, M.A.E.; Tork, O.M.; Rashed, L.A.; Ibrahim, W.; Morsi, H.; Rabie, D.M.M. Mitochondrial dysfunction in diabetic cardiomyopathy: Effect of mesenchymal stem cell with PPAR-gamma agonist or exendin-4. Exp. Clin. Endocrinol. Diabetes 2018, 126, 27–38. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, J.; Xu, S.; Zhou, W.; Wu, L.; Wang, L.; Li, W. Exendin-4 reduces ventricular arrhythmia activity and calcium sparks-mediated sarcoplasmic reticulum calcium leak in rats with heart failure. Int. Heart J. 2020, 61, 145–152. [Google Scholar] [CrossRef] [Green Version]
  85. Piao, L.; Zhao, G.; Zhu, E.; Inoue, A.; Shibata, R.; Lei, Y.; Hu, L.; Yu, C.; Yang, G.; Wu, H.; et al. Chronic psychological stress accelerates vascular senescence and impairs ischemia-induced neovascularization: The role of dipeptidyl peptidase-4/glucagon-like peptide-1-adiponectin axis. J. Am. Heart Assoc. 2017, 6, e006421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Katsurada, K.; Nakata, M.; Saito, T.; Zhang, B.; Maejima, Y.; Nandi, S.S.; Sharma, N.M.; Patel, K.P.; Kario, K.; Yada, T. Central glucagon-like peptide-1 receptor signaling via brainstem catecholamine neurons counteracts hypertension in spontaneously hypertensive rats. Sci. Rep. 2019, 9, 12986. [Google Scholar] [CrossRef] [Green Version]
  87. Jojima, T.; Uchida, K.; Akimoto, K.; Tomotsune, T.; Yanagi, K.; Iijima, T.; Suzuki, K.; Kasai, K.; Aso, Y. Liraglutide, a GLP-1 receptor agonist, inhibits vascular smooth muscle cell proliferation by enhancing AMP-activated protein kinase and cell cycle regulation, and delays atherosclerosis in ApoE deficient mice. Atherosclerosis 2017, 261, 44–51. [Google Scholar] [CrossRef]
  88. Zhang, Z.; Wang, X.; Yang, L.; Yang, L.; Ma, H. Liraglutide ameliorates myocardial damage in experimental diabetic rats by inhibiting pyroptosis via Sirt1/AMPK signaling. Iran. J. Basic Med. Sci. 2021, 24, 1358–1365. [Google Scholar] [CrossRef]
  89. Zheng, R.H.; Zhang, W.W.; Ji, Y.N.; Bai, X.J.; Yan, C.P.; Wang, J.; Bai, F.; Zhao, Z.Q. Exogenous supplement of glucagon like peptide-1 protects the heart against aortic banding induced myocardial fibrosis and dysfunction through inhibiting mTOR/p70S6K signaling and promoting autophagy. Eur. J. Pharmacol. 2020, 883, 173318. [Google Scholar] [CrossRef]
  90. Scheen, A.J. GLP-1 receptor agonists and heart failure in diabetes. Diabetes Metab. 2017, 43 (Suppl. S1), 2S13–2S19. [Google Scholar] [CrossRef]
  91. Sokos, G.G.; Nikolaidis, L.A.; Mankad, S.; Elahi, D.; Shannon, R.P. Glucagon-like peptide-1 infusion improves left ventricular ejection fraction and functional status in patients with chronic heart failure. J. Card. Fail. 2006, 12, 694–699. [Google Scholar] [CrossRef] [PubMed]
  92. Lepore, J.J.; Olson, E.; Demopoulos, L.; Haws, T.; Fang, Z.; Barbour, A.M.; Fossler, M.; Davila-Roman, V.G.; Russell, S.D.; Gropler, R.J. Effects of the novel long-acting GLP-1 agonist, albiglutide, on cardiac function, cardiac metabolism, and exercise capacity in patients with chronic heart failure and reduced ejection fraction. JACC Heart Fail. 2016, 4, 559–566. [Google Scholar] [CrossRef] [PubMed]
  93. Scalzo, R.L.; Moreau, K.L.; Ozemek, C.; Herlache, L.; McMillin, S.; Gilligan, S.; Huebschmann, A.G.; Bauer, T.A.; Dorosz, J.; Reusch, J.E.; et al. Exenatide improves diastolic function and attenuates arterial stiffness but does not alter exercise capacity in individuals with type 2 diabetes. J. Diabetes Complicat. 2017, 31, 449–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Baggio, L.L.; Ussher, J.R.; McLean, B.A.; Cao, X.; Kabir, M.G.; Mulvihill, E.E.; Mighiu, A.S.; Zhang, H.; Ludwig, A.; Seeley, R.J.; et al. The autonomic nervous system and cardiac GLP-1 receptors control heart rate in mice. Mol. Metab. 2017, 6, 1339–1349. [Google Scholar] [CrossRef] [PubMed]
  95. Yamamoto, H.; Lee, C.E.; Marcus, J.N.; Williams, T.D.; Overton, J.M.; Lopez, M.E.; Hollenberg, A.N.; Baggio, L.; Saper, C.B.; Drucker, D.J.; et al. Glucagon-like peptide-1 receptor stimulation increases blood pressure and heart rate and activates autonomic regulatory neurons. J. Clin. Investig. 2002, 110, 43–52. [Google Scholar] [CrossRef]
  96. Ang, R.; Mastitskaya, S.; Hosford, P.S.; Basalay, M.; Specterman, M.; Aziz, Q.; Li, Y.; Orini, M.; Taggart, P.; Lambiase, P.D.; et al. Modulation of cardiac ventricular excitability by GLP-1 (glucagon-like peptide-1). Circ. Arrhythm. Electrophysiol. 2018, 11, e006740. [Google Scholar] [CrossRef] [PubMed]
  97. Barragan, J.M.; Eng, J.; Rodriguez, R.; Blazquez, E. Neural contribution to the effect of glucagon-like peptide-1-(7-36) amide on arterial blood pressure in rats. Am. J. Physiol. 1999, 277, E784–E791. [Google Scholar] [CrossRef]
  98. Barragan, J.M.; Rodriguez, R.E.; Blazquez, E. Changes in arterial blood pressure and heart rate induced by glucagon-like peptide-1-(7-36) amide in rats. Am. J. Physiol. 1994, 266, E459–E466. [Google Scholar] [CrossRef]
  99. Okerson, T.; Yan, P.; Stonehouse, A.; Brodows, R. Effects of exenatide on systolic blood pressure in subjects with type 2 diabetes. Am. J. Hypertens. 2010, 23, 334–339. [Google Scholar] [CrossRef] [Green Version]
  100. Blonde, L.; Russell-Jones, D. The safety and efficacy of liraglutide with or without oral antidiabetic drug therapy in type 2 diabetes: An overview of the LEAD 1-5 studies. Diabetes Obes. Metab. 2009, 11 (Suppl. S3), 26–34. [Google Scholar] [CrossRef]
  101. Klonoff, D.C.; Buse, J.B.; Nielsen, L.L.; Guan, X.; Bowlus, C.L.; Holcombe, J.H.; Wintle, M.E.; Maggs, D.G. Exenatide effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type 2 diabetes treated for at least 3 years. Curr. Med. Res. Opin. 2008, 24, 275–286. [Google Scholar] [CrossRef]
  102. Ravassa, S.; Zudaire, A.; Diez, J. GLP-1 and cardioprotection: From bench to bedside. Cardiovasc. Res. 2012, 94, 316–323. [Google Scholar] [CrossRef] [Green Version]
  103. Wei, Y.; Mojsov, S. Tissue-specific expression of the human receptor for glucagon-like peptide-I: Brain, heart and pancreatic forms have the same deduced amino acid sequences. FEBS Lett. 1995, 358, 219–224. [Google Scholar] [CrossRef] [Green Version]
  104. Kim, M.; Platt, M.J.; Shibasaki, T.; Quaggin, S.E.; Backx, P.H.; Seino, S.; Simpson, J.A.; Drucker, D.J. GLP-1 receptor activation and Epac2 link atrial natriuretic peptide secretion to control of blood pressure. Nat. Med. 2013, 19, 567–575. [Google Scholar] [CrossRef]
  105. Rudovich, N.; Pivovarova, O.; Gogebakan, O.; Sparwasser, A.; Doehner, W.; Anker, S.D.; Arafat, A.M.; Bergmann, A.; Nauck, M.A.; Pfeiffer, A.F. Effect of exogenous intravenous administrations of GLP-1 and/or GIP on circulating pro-atrial natriuretic peptide in subjects with different stages of glucose tolerance. Diabetes Care 2015, 38, e7–e8. [Google Scholar] [CrossRef] [Green Version]
  106. Zhao, T.; Parikh, P.; Bhashyam, S.; Bolukoglu, H.; Poornima, I.; Shen, Y.T.; Shannon, R.P. Direct effects of glucagon-like peptide-1 on myocardial contractility and glucose uptake in normal and postischemic isolated rat hearts. J. Pharmacol. Exp. Ther. 2006, 317, 1106–1113. [Google Scholar] [CrossRef]
  107. Lu, K.; Chang, G.; Ye, L.; Zhang, P.; Li, Y.; Zhang, D. Protective effects of extendin-4 on hypoxia/reoxygenation-induced injury in H9c2 cells. Mol. Med. Rep. 2015, 12, 3007–3016. [Google Scholar] [CrossRef] [Green Version]
  108. Gejl, M.; Søndergaard, H.M.; Stecher, C.; Bibby, B.M.; Møller, N.; Bøtker, H.E.; Hansen, S.B.; Gjedde, A.; Rungby, J.; Brock, B. Exenatide alters myocardial glucose transport and uptake depending on insulin resistance and increases myocardial blood flow in patients with type 2 diabetes. J. Clin. Endocrinol. Metab. 2012, 97, E1165–E1169. [Google Scholar] [CrossRef]
  109. Gejl, M.; Lerche, S.; Mengel, A.; Møller, N.; Bibby, B.M.; Smidt, K.; Brock, B.; Søndergaard, H.; Bøtker, H.E.; Gjedde, A.; et al. Influence of GLP-1 on myocardial glucose metabolism in healthy men during normo- or hypoglycemia. PLoS ONE 2014, 9, e83758. [Google Scholar] [CrossRef]
  110. Ying, Y.; Zhu, H.; Liang, Z.; Ma, X.; Li, S. GLP1 protects cardiomyocytes from palmitate-induced apoptosis via Akt/GSK3b/b-catenin pathway. J. Mol. Endocrinol. 2015, 55, 245–262. [Google Scholar] [CrossRef] [Green Version]
  111. Sonne, D.P.; Engstrom, T.; Treiman, M. Protective effects of GLP-1 analogues exendin-4 and GLP-1(9-36) amide against ischemia-reperfusion injury in rat heart. Regul. Pept. 2008, 146, 243–249. [Google Scholar] [CrossRef]
  112. Timmers, L.; Henriques, J.P.; de Kleijn, D.P.; Devries, J.H.; Kemperman, H.; Steendijk, P.; Verlaan, C.W.; Kerver, M.; Piek, J.J.; Doevendans, P.A.; et al. Exenatide reduces infarct size and improves cardiac function in a porcine model of ischemia and reperfusion injury. J. Am. Coll. Cardiol. 2009, 53, 501–510. [Google Scholar] [CrossRef] [Green Version]
  113. Nikolaidis, L.A.; Mankad, S.; Sokos, G.G.; Miske, G.; Shah, A.; Elahi, D.; Shannon, R.P. Effects of glucagon-like peptide-1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusion. Circulation 2004, 109, 962–965. [Google Scholar] [CrossRef] [Green Version]
  114. Guan, G.; Zhang, J.; Liu, S.; Huang, W.; Gong, Y.; Gu, X. Glucagon-like peptide-1 attenuates endoplasmic reticulum stress-induced apoptosis in H9c2 cardiomyocytes during hypoxia/reoxygenation through the GLP-1R/PI3K/Akt pathways. Naunyn. Schmiedebergs Arch. Pharmacol. 2019, 392, 715–722. [Google Scholar] [CrossRef]
  115. Ban, K.; Kim, K.H.; Cho, C.K.; Sauve, M.; Diamandis, E.P.; Backx, P.H.; Drucker, D.J.; Husain, M. Glucagon-like peptide (GLP)-1(9-36)amide-mediated cytoprotection is blocked by exendin(9-39) yet does not require the known GLP-1 receptor. Endocrinology 2010, 151, 1520–1531. [Google Scholar] [CrossRef] [Green Version]
  116. Noyan-Ashraf, M.H.; Momen, M.A.; Ban, K.; Sadi, A.M.; Zhou, Y.Q.; Riazi, A.M.; Baggio, L.L.; Henkelman, R.M.; Husain, M.; Drucker, D.J. GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes 2009, 58, 975–983. [Google Scholar] [CrossRef] [Green Version]
  117. Maack, C.; Kartes, T.; Kilter, H.; Schafers, H.J.; Nickenig, G.; Bohm, M.; Laufs, U. Oxygen free radical release in human failing myocardium is associated with increased activity of rac1-GTPase and represents a target for statin treatment. Circulation 2003, 108, 1567–1574. [Google Scholar] [CrossRef] [Green Version]
  118. Fukui, T.; Yoshiyama, M.; Hanatani, A.; Omura, T.; Yoshikawa, J.; Abe, Y. Expression of p22-phox and gp91-phox, essential components of NADPH oxidase, increases after myocardial infarction. Biochem. Biophys. Res. Commun. 2001, 281, 1200–1206. [Google Scholar] [CrossRef]
  119. Mangmool, S.; Denkaew, T.; Phosri, S.; Pinthong, D.; Parichatikanond, W.; Shimauchi, T.; Nishida, M. Sustained betaAR stimulation mediates cardiac insulin resistance in a PKA-dependent manner. Mol. Endocrinol. 2016, 30, 118–132. [Google Scholar] [CrossRef] [Green Version]
  120. Parichatikanond, W.; Nishimura, A.; Nishida, M.; Mangmool, S. Prolonged stimulation of beta(2)-adrenergic receptor with beta(2)-agonists impairs insulin actions in H9c2 cells. J. Pharmacol. Sci. 2018, 138, 184–191. [Google Scholar] [CrossRef]
  121. Hutchinson, D.S.; Chernogubova, E.; Dallner, O.S.; Cannon, B.; Bengtsson, T. Beta-adrenoceptors, but not alpha-adrenoceptors, stimulate AMP-activated protein kinase in brown adipocytes independently of uncoupling protein-1. Diabetologia 2005, 48, 2386–2395. [Google Scholar] [CrossRef] [Green Version]
  122. Chiasson, J.L.; Shikama, H.; Chu, D.T.; Exton, J.H. Inhibitory effect of epinephrine on insulin-stimulated glucose uptake by rat skeletal muscle. J. Clin. Investig. 1981, 68, 706–713. [Google Scholar] [CrossRef] [Green Version]
  123. Ban, K.; Noyan-Ashraf, M.H.; Hoefer, J.; Bolz, S.S.; Drucker, D.J.; Husain, M. Cardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation 2008, 117, 2340–2350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Ma, T.; Du, X.; Pick, J.E.; Sui, G.; Brownlee, M.; Klann, E. Glucagon-like peptide-1 cleavage product GLP-1(9-36) amide rescues synaptic plasticity and memory deficits in Alzheimer’s disease model mice. J. Neurosci. 2012, 32, 13701–13708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Vakifahmetoglu-Norberg, H.; Ouchida, A.T.; Norberg, E. The role of mitochondria in metabolism and cell death. Biochem. Biophys. Res. Commun. 2017, 482, 426–431. [Google Scholar] [CrossRef]
  126. Volpe, C.M.O.; Villar-Delfino, P.H.; Dos Anjos, P.M.F.; Nogueira-Machado, J.A. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018, 9, 119. [Google Scholar] [CrossRef] [Green Version]
  127. Marso, S.P.; Bain, S.C.; Consoli, A.; Eliaschewitz, F.G.; Jodar, E.; Leiter, L.A.; Lingvay, I.; Rosenstock, J.; Seufert, J.; Warren, M.L.; et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N. Engl. J. Med. 2016, 375, 1834–1844. [Google Scholar] [CrossRef] [Green Version]
  128. Marso, S.P.; Daniels, G.H.; Brown-Frandsen, K.; Kristensen, P.; Mann, J.F.; Nauck, M.A.; Nissen, S.E.; Pocock, S.; Poulter, N.R.; Ravn, L.S.; et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N. Engl. J. Med. 2016, 375, 311–322. [Google Scholar] [CrossRef] [Green Version]
  129. Kyriacou, A.; Ahmed, A.B. Exenatide use in the management of type 2 diabetes mellitus. Pharmaceuticals 2010, 3, 2554–2567. [Google Scholar] [CrossRef] [Green Version]
  130. Shyangdan, D.; Cummins, E.; Royle, P.; Waugh, N. Liraglutide for the treatment of type 2 diabetes. Health Technol. Assess. 2011, 15 (Suppl. S1), 77–86. [Google Scholar] [CrossRef]
  131. Nauck, M.A.; Friedrich, N. Do GLP-1-based therapies increase cancer risk? Diabetes Care 2013, 36 (Suppl. S2), S245–S252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Vangoitsenhoven, R.; Mathieu, C.; Van der Schueren, B. GLP1 and cancer: Friend or foe? Endocr. Relat. Cancer 2012, 19, F77–F88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Chang, G.; Liu, J.; Qin, S.; Jiang, Y.; Zhang, P.; Yu, H.; Lu, K.; Zhang, N.; Cao, L.; Wang, Y.; et al. Cardioprotection by exenatide: A novel mechanism via improving mitochondrial function involving the GLP-1 receptor/cAMP/PKA pathway. Int. J. Mol. Med. 2018, 41, 1693–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Zhang, L.; Tian, J.; Diao, S.; Zhang, G.; Xiao, M.; Chang, D. GLP-1 receptor agonist liraglutide protects cardiomyocytes from IL-1beta-induced metabolic disturbance and mitochondrial dysfunction. Chem. Biol. Interact. 2020, 332, 109252. [Google Scholar] [CrossRef]
  135. Yuan, M.; Gong, M.; Zhang, Z.; Meng, L.; Tse, G.; Zhao, Y.; Bao, Q.; Zhang, Y.; Yuan, M.; Liu, X.; et al. Hyperglycemia induces endoplasmic reticulum stress in atrial cardiomyocytes, and mitofusin-2 downregulation prevents mitochondrial dysfunction and subsequent cell death. Oxidative Med. Cell. Longev. 2020, 2020, 6569728. [Google Scholar] [CrossRef]
  136. Labbadia, J.; Morimoto, R.I. The biology of proteostasis in aging and disease. Annu. Rev. Biochem. 2015, 84, 435–464. [Google Scholar] [CrossRef] [Green Version]
  137. Sabath, N.; Levy-Adam, F.; Younis, A.; Rozales, K.; Meller, A.; Hadar, S.; Soueid-Baumgarten, S.; Shalgi, R. Cellular proteostasis decline in human senescence. Proc. Natl. Acad. Sci. USA 2020, 117, 31902–31913. [Google Scholar] [CrossRef]
  138. DeNicola, M.; Du, J.; Wang, Z.; Yano, N.; Zhang, L.; Wang, Y.; Qin, G.; Zhuang, S.; Zhao, T.C. Stimulation of glucagon-like peptide-1 receptor through exendin-4 preserves myocardial performance and prevents cardiac remodeling in infarcted myocardium. Am. J. Physiol. Endocrinol. Metab. 2014, 307, E630–E643. [Google Scholar] [CrossRef] [Green Version]
  139. Qiao, H.; Ren, H.; Du, H.; Zhang, M.; Xiong, X.; Lv, R. Liraglutide repairs the infarcted heart: The role of the SIRT1/Parkin/mitophagy pathway. Mol. Med. Rep. 2018, 17, 3722–3734. [Google Scholar] [CrossRef] [Green Version]
  140. Kyhl, K.; Lonborg, J.; Hartmann, B.; Kissow, H.; Poulsen, S.S.; Ali, H.E.; Kjaer, A.; Dela, F.; Engstrom, T.; Treiman, M. Lack of effect of prolonged treatment with liraglutide on cardiac remodeling in rats after acute myocardial infarction. Peptides 2017, 93, 1–12. [Google Scholar] [CrossRef]
  141. Yu, W.; Zha, W.; Ren, J. Exendin-4 and liraglutide attenuate glucose toxicity-induced cardiac injury through mTOR/ULK1-dependent autophagy. Oxidative Med. Cell. Longev. 2018, 2018, 5396806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Ludwig, D.S.; Apovian, C.M.; Aronne, L.J.; Astrup, A.; Cantley, L.C.; Ebbeling, C.B.; Heymsfield, S.B.; Johnson, J.D.; King, J.C.; Krauss, R.M.; et al. Competing paradigms of obesity pathogenesis: Energy balance versus carbohydrate-insulin models. Eur. J. Clin. Nutr. 2022, 76, 1209–1221. [Google Scholar] [CrossRef] [PubMed]
  143. Finan, B.; Ma, T.; Ottaway, N.; Muller, T.D.; Habegger, K.M.; Heppner, K.M.; Kirchner, H.; Holland, J.; Hembree, J.; Raver, C.; et al. Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans. Sci. Transl. Med. 2013, 5, 209ra151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Adriaenssens, A.E.; Gribble, F.M.; Reimann, F. The glucose-dependent insulinotropic polypeptide signaling axis in the central nervous system. Peptides 2020, 125, 170194. [Google Scholar] [CrossRef]
  145. NamKoong, C.; Kim, M.S.; Jang, B.T.; Lee, Y.H.; Cho, Y.M.; Choi, H.J. Central administration of GLP-1 and GIP decreases feeding in mice. Biochem. Biophys. Res. Commun. 2017, 490, 247–252. [Google Scholar] [CrossRef]
  146. Zhang, Q.; Delessa, C.T.; Augustin, R.; Bakhti, M.; Collden, G.; Drucker, D.J.; Feuchtinger, A.; Caceres, C.G.; Grandl, G.; Harger, A.; et al. The glucose-dependent insulinotropic polypeptide (GIP) regulates body weight and food intake via CNS-GIPR signaling. Cell Metab. 2021, 33, 833–844.e5. [Google Scholar] [CrossRef]
  147. Nauck, M.A.; D’Alessio, D.A. Tirzepatide, a dual GIP/GLP-1 receptor co-agonist for the treatment of type 2 diabetes with unmatched effectiveness regrading glycaemic control and body weight reduction. Cardiovasc. Diabetol. 2022, 21, 169. [Google Scholar] [CrossRef]
  148. Fisman, E.Z.; Tenenbaum, A. The dual glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) receptor agonist tirzepatide: A novel cardiometabolic therapeutic prospect. Cardiovasc. Diabetol. 2021, 20, 225. [Google Scholar] [CrossRef]
  149. Frias, J.P.; Nauck, M.A.; Van, J.; Kutner, M.E.; Cui, X.; Benson, C.; Urva, S.; Gimeno, R.E.; Milicevic, Z.; Robins, D.; et al. Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: A randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet 2018, 392, 2180–2193. [Google Scholar] [CrossRef]
  150. Pirro, V.; Roth, K.D.; Lin, Y.; Willency, J.A.; Milligan, P.L.; Wilson, J.M.; Ruotolo, G.; Haupt, A.; Newgard, C.B.; Duffin, K.L. Effects of tirzepatide, a dual GIP and GLP-1 RA, on lipid and metabolite profiles in subjects with type 2 diabetes. J. Clin. Endocrinol. Metab. 2022, 107, 363–378. [Google Scholar] [CrossRef]
  151. Wilson, J.M.; Lin, Y.; Luo, M.J.; Considine, G.; Cox, A.L.; Bowsman, L.M.; Robins, D.A.; Haupt, A.; Duffin, K.L.; Ruotolo, G. The dual glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1 receptor agonist tirzepatide improves cardiovascular risk biomarkers in patients with type 2 diabetes: A post hoc analysis. Diabetes Obes. Metab. 2022, 24, 148–153. [Google Scholar] [CrossRef] [PubMed]
  152. Wilson, J.M.; Nikooienejad, A.; Robins, D.A.; Roell, W.C.; Riesmeyer, J.S.; Haupt, A.; Duffin, K.L.; Taskinen, M.R.; Ruotolo, G. The dual glucose-dependent insulinotropic peptide and glucagon-like peptide-1 receptor agonist, tirzepatide, improves lipoprotein biomarkers associated with insulin resistance and cardiovascular risk in patients with type 2 diabetes. Diabetes Obes. Metab. 2020, 22, 2451–2459. [Google Scholar] [CrossRef] [PubMed]
  153. Sattar, N.; McGuire, D.K.; Pavo, I.; Weerakkody, G.J.; Nishiyama, H.; Wiese, R.J.; Zoungas, S. Tirzepatide cardiovascular event risk assessment: A pre-specified meta-analysis. Nat. Med. 2022, 28, 591–598. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structural arrangement of mammalian proglucagon. (A) The amino acid sequences in proglucagon have been marked as numbers. The post-translational processing produces peptides in the intestine and pancreas as shown. (B) The DPP-4 cleavage site in the amino acid chain is indicated by arrows. GRPP, glicentin-related pancreatic polypeptide; IP, intervening peptide; PC1/2/3, prohormone convertases 1/2/3; DPP-4, dipeptidyl peptidase-4.
Figure 1. Structural arrangement of mammalian proglucagon. (A) The amino acid sequences in proglucagon have been marked as numbers. The post-translational processing produces peptides in the intestine and pancreas as shown. (B) The DPP-4 cleavage site in the amino acid chain is indicated by arrows. GRPP, glicentin-related pancreatic polypeptide; IP, intervening peptide; PC1/2/3, prohormone convertases 1/2/3; DPP-4, dipeptidyl peptidase-4.
Pharmaceuticals 16 00836 g001
Figure 2. GLP-1 receptor (GLP-1R) gene expression (transcripts per million; TPM) amongst females and males in selected tissues related to the cardiovascular system and pancreas (from GTEx Portal https://gtexportal.org/home/gene/GLP-1R accessed on 21 November 2022). Box plots are depicted as median and correspond to 25th and 75th percentiles. The pancreas expresses the higher expression (median value, 4.5) when compared to heart-atria (median value, 1.7), heart left ventricle (median value, 0.6), coronary artery (median value, 0.05), and aorta (median value, not available). In this cohort analysis, there are no significant sex differences in the expression pattern of GLP-1R amongst males and females.
Figure 2. GLP-1 receptor (GLP-1R) gene expression (transcripts per million; TPM) amongst females and males in selected tissues related to the cardiovascular system and pancreas (from GTEx Portal https://gtexportal.org/home/gene/GLP-1R accessed on 21 November 2022). Box plots are depicted as median and correspond to 25th and 75th percentiles. The pancreas expresses the higher expression (median value, 4.5) when compared to heart-atria (median value, 1.7), heart left ventricle (median value, 0.6), coronary artery (median value, 0.05), and aorta (median value, not available). In this cohort analysis, there are no significant sex differences in the expression pattern of GLP-1R amongst males and females.
Pharmaceuticals 16 00836 g002
Figure 3. Systemic effects of GLP-1 and its analogs. The effects on various tissues/organs illustrated in the figure are based on results obtained in several preclinical or clinical studies at physiological GLP-1 levels or after exposure to GLP-1R analogs. GLP-1R is abundantly distributed in the tissues/organs of the peripheral and central nervous system with the different expression levels leading to the distinct biological functions in each tissue/organ. In the CNS and pancreas, high expression of GLP-1R is detected, while lower expression of GLP-1R is observed in the heart, lungs, gut, muscle, kidneys, liver, PNS, and other tissues. ↑, Increase; ↓, Decrease.
Figure 3. Systemic effects of GLP-1 and its analogs. The effects on various tissues/organs illustrated in the figure are based on results obtained in several preclinical or clinical studies at physiological GLP-1 levels or after exposure to GLP-1R analogs. GLP-1R is abundantly distributed in the tissues/organs of the peripheral and central nervous system with the different expression levels leading to the distinct biological functions in each tissue/organ. In the CNS and pancreas, high expression of GLP-1R is detected, while lower expression of GLP-1R is observed in the heart, lungs, gut, muscle, kidneys, liver, PNS, and other tissues. ↑, Increase; ↓, Decrease.
Pharmaceuticals 16 00836 g003
Figure 4. Schematic diagram representing cardioprotective molecular mechanisms of GLP-1 analogs in cardiomyocytes. The GLP-1R-dependent and -independent signaling pathways have been described as contributing to healthy myocardium and heart functions under pathophysiological conditions. The binding of GLP-1 to its receptor activates adenylate cyclase, and cAMP levels are elevated, which is followed by the activation of downstream cAMP sensitive molecular effectors in a GLP-1R-dependent way. While GLP-1R-independent GLP-1(9-36) activates PI3K, Akt, and the downstream signaling pathway, the full mechanisms of this process are not yet known. GLP-1, glucagon-like peptide-1; cAMP, cyclic adenosine monophosphate; Akt, protein kinase B; Epac, exchange protein activated by cAMP; CREB, cAMP-response element binding protein; CaMKKβ, calcium ions/calmodulins dependent protein kinase kinases β; PINK1, PTEN-induced kinase 1; Mfn, Mitofusin; Drp, dynamin-related protein; NF-κB, nuclear factor-κB; eNOS, endothelial nitric oxide synthase; AMPK, AMP-activated protein kinase; mTOR, mammalian target of rapamycin; ERK, extracellular-signal-regulated kinase; PI3K, phosphatidylinositide 3-kinase.
Figure 4. Schematic diagram representing cardioprotective molecular mechanisms of GLP-1 analogs in cardiomyocytes. The GLP-1R-dependent and -independent signaling pathways have been described as contributing to healthy myocardium and heart functions under pathophysiological conditions. The binding of GLP-1 to its receptor activates adenylate cyclase, and cAMP levels are elevated, which is followed by the activation of downstream cAMP sensitive molecular effectors in a GLP-1R-dependent way. While GLP-1R-independent GLP-1(9-36) activates PI3K, Akt, and the downstream signaling pathway, the full mechanisms of this process are not yet known. GLP-1, glucagon-like peptide-1; cAMP, cyclic adenosine monophosphate; Akt, protein kinase B; Epac, exchange protein activated by cAMP; CREB, cAMP-response element binding protein; CaMKKβ, calcium ions/calmodulins dependent protein kinase kinases β; PINK1, PTEN-induced kinase 1; Mfn, Mitofusin; Drp, dynamin-related protein; NF-κB, nuclear factor-κB; eNOS, endothelial nitric oxide synthase; AMPK, AMP-activated protein kinase; mTOR, mammalian target of rapamycin; ERK, extracellular-signal-regulated kinase; PI3K, phosphatidylinositide 3-kinase.
Pharmaceuticals 16 00836 g004
Table 1. List of current GLP-1 and its analogs [36,37,38,39].
Table 1. List of current GLP-1 and its analogs [36,37,38,39].
GLP-1 AnalogsCompanyFDA Approval Year for T2DMDescriptionAdministrationHalf-Life
Human GLP-1 backbone (_glutide)
AlbiglutideGlaxoSmithKline2014Extended release,
fused to human albumin
Subcutaneous (SC), once weekly5–6 days
DulaglutideEli Lilly2014Extended release,
Fc region of human IgG4
SC, once weekly4.7 days
LiraglutideNovo Nordisk2010Immediate release,
linked with free fatty acid
SC, once daily12–13 h
SemaglutideNovo Nordisk2017Extended release,
linked with free fatty acid
SC, once weekly5–7 days
Novo Nordisk2020Extended release,
linked with free fatty acid
Oral, once daily5–7 days
Exendin-4 backbone (_natide)
ExenatideEli Lilly2005Immediate releaseSC, twice daily2.4 h
AstraZeneca2012Extended release, encapsulated in microsphere formSC, once weekly3–5 days
EfpeglenatideSanofiNot availableExtended releaseSC, once monthlyNot available
LixisenatideSanofi2016Immediate release,
linked with poly-lysine tail
SC, once daily3–4 h
Table 2. Effects of GLP-1 and its analogs on organs/tissues [38,44,45].
Table 2. Effects of GLP-1 and its analogs on organs/tissues [38,44,45].
Organs/TissuesBiological Effects
Heart
Protect against cardiac ischemia and myocardial damage;
Enhance heart rate and cardiac output;
Improve glucose utilization and increase fatty acid metabolism;
Vasodilation and anti-inflammatory effect;
Increase atrial natriuretic peptide (ANP) secretion.
Brain
Suppress appetite and increase satiety;
Neuroprotection, improve learning and memory;
Induce proliferation of neural stem cells;
Decrease inflammation.
Liver
Decrease glucose production;
Reduce liver fat and steatosis.
Kidneys
Diuresis and natriuresis.
Pancreas
Enhance somatostatin and insulin secretion;
Inhibit apoptosis and increase proliferation of β-cells.
Gut
Delay gastric emptying time and acid secretion;
Inhibit peristalsis and lipoprotein secretion.
Adipose tissues
Increase lipolysis and glucose uptake.
Immune system
Reduce inflammation.
Skeletal muscle
Induce glucose uptake and tissue perfusion.
Reproductive
system
Improve ovarian function;
Increase menstrual frequency and ovulation rates;
Decrease infertility.
Table 3. Preclinical studies of GLP-1R agonists for cardioprotective effects (In vitro).
Table 3. Preclinical studies of GLP-1R agonists for cardioprotective effects (In vitro).
GLP-1 AnalogsStudy ModelsMechanistic FindingsRef.
Exendin-4Methylglyoxal-exposed
H9c2 cardiomyoblasts
Inhibits intracellular and mitochondrial ROS production, apoptosis, and MMP impairment;
Upregulates genes related to mitochondrial functions and dynamics via cAMP/Epac/PI3K/Akt pathway.
[70]
Exendin-4Hydrogen peroxide (H2O2) -exposed neonatal rat cardiomyocytes
Exhibits antioxidant effects by inducing the synthesis of catalase, GPx-1, and Mn-SOD via Epac signaling;
Elicits anti-apoptotic effects by enhancing Bcl-2 expression and inhibiting caspase-3 activity via PKA and Epac signaling.
[71]
Exendin-4Phenylephrine-induced
neonatal rat cardiomyocyte
Attenuates cardiac hypertrophy by upregulating GLP-1R expression and activating AMPK/mTOR signaling.
[72]
Exendin-4Oxygen glucose deprivation/reoxygenation (OGD/R)
in human ventricular cardiomyocytes
Restores autophagic flux induced by OGD/R through promoting nuclear translocation of TFEB;
Reduces infarct size and preserves cardiac function through anti-apoptosis and antioxidative effects.
[73]
Exendin-4High-density lipoprotein (HDL)-induced human umbilical vein endothelial cells
Protects endothelial function by increasing HDL scavenger receptor class BI expression via AMPK/FoxO1 pathway and activating eNOS.
[74]
GLP-1HL-1 cells derived from mouse atrial cardiac muscle cells
Regulates arrhythmogenesis through modulating calcium handling proteins.
[75]
LiraglutideAngiotensin II-treated H9c2 cells
Decreases cardiac hypertrophy by increasing GLP-1R expression and activating AMPK;
Reduces the expression of AngII/AT1R/ACE2 and mTOR.
[76]
SemaglutideLipopolysaccharides-treated H9c2 cells
Protects exercise-induced cardiomyopathy by activating AMPK pathway;
Increases autophagy, reduces ROS production and inflammatory markers (NF-κB, TNF-α, and IL-1β).
[77]
SemaglutideHypoxia/reoxygenation (H/R) injury in H9c2 cells
Inhibits H/R injury-induced cardiomyocyte apoptosis by activating PKG/PKCε/ERK1/2 pathway.
[78]
Table 4. Preclinical studies of GLP-1R agonists for cardioprotective effects (In vivo).
Table 4. Preclinical studies of GLP-1R agonists for cardioprotective effects (In vivo).
GLP-1 AnalogsStudy ModelsMechanistic FindingsRef.
Exendin-4Left anterior descending (LAD) coronary artery ligation-induced heart failure rats
Attenuates cardiac remodeling through activation of eNOS/cGMP/PKG pathway.
[79]
Exendin-4LAD coronary artery ligation-induced myocardial infarction (MI) rats
Inhibits collagen I, collagen III, transforming growth factor-β1, phosphorylated PI3K, and Akt levels.
[80]
Exendin-4High-fat diet (HFD)-induced diabetic rats
Inhibits cardiomyocyte pyroptosis via AMPK-TXNIP pathway.
[81]
Exendin-4LAD coronary artery ligation-induced MI rats
Attenuates myocardial remodeling after an acute MI by activating β-arrestin-2, protein phosphatase 2A, and GSK-3, and inhibiting β-catenin.
[82]
Exendin-4Streptozotocin (STZ)-induced diabetic rats
Downregulates NF-kB and upregulates cardiolipin, PGC1α, LC3, and beclin.
[83]
Exendin-4LAD coronary artery ligation-induced MI rats
Reduces ventricular arrhythmias through decreasing SR calcium leak, reduced RyR2 phosphorylation, and decreased CaMK-II activity.
[84]
ExenatideDPP4-deficient rats;
Adiponectin-deficient mice
Prevents chronic stress induced vascular senescence.
[85]
Exendin-4LAD coronary artery ligation-induced MI rats
Decreases susceptibility to atrial arrhythmogenesis;
Improves conduction properties and exerts antifibrotic effects via GLP-1R signaling.
[80]
Exendin-4LAD coronary artery ligation-induced MI rats
Increases GLP-1/GLP-1R expression in ventricular tissues;
Activates eNOS/cGMP/PKG signaling and inhibits CaMKII pathway;
Inhibits pathological cardiac remodeling;
[79]
LiraglutideSpontaneously hypertensive rats (SHR)
Delays progression of hypertension through activation of brainstem dopamine beta-hydroxylase (DBH) neurons;
Suppresses sympathetic nerve activity.
[86]
LiraglutideApolipoprotein E deficient (ApoE−/−) mice
Suppresses atherosclerotic lesions;
Increases AMPK phosphorylation in aortic wall.
[87]
LiraglutideHFD-feeding and STZ-induced diabetic rats
Reduces myocardial damage by activating the Sirt1/AMPK pathways;
Inhibits cellular pyroptosis.
[88]
LiraglutideAbdominal aortic constriction-induced cardiac fibrosis rats
Inhibits cardiac fibrosis and dysfunction by inhibiting mTOR/p70S6K signaling and enhancing autophagy.
[89]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pandey, S.; Mangmool, S.; Parichatikanond, W. Multifaceted Roles of GLP-1 and Its Analogs: A Review on Molecular Mechanisms with a Cardiotherapeutic Perspective. Pharmaceuticals 2023, 16, 836. https://doi.org/10.3390/ph16060836

AMA Style

Pandey S, Mangmool S, Parichatikanond W. Multifaceted Roles of GLP-1 and Its Analogs: A Review on Molecular Mechanisms with a Cardiotherapeutic Perspective. Pharmaceuticals. 2023; 16(6):836. https://doi.org/10.3390/ph16060836

Chicago/Turabian Style

Pandey, Sudhir, Supachoke Mangmool, and Warisara Parichatikanond. 2023. "Multifaceted Roles of GLP-1 and Its Analogs: A Review on Molecular Mechanisms with a Cardiotherapeutic Perspective" Pharmaceuticals 16, no. 6: 836. https://doi.org/10.3390/ph16060836

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop