Next Article in Journal
Involvement of Cellular Prion Protein in Invasion and Metastasis of Lung Cancer by Inducing Treg Cell Development
Next Article in Special Issue
Glucagon Prevents Cytotoxicity Induced by Methylglyoxal in a Rat Neuronal Cell Line Model
Previous Article in Journal
The Interrelationships between Intestinal Permeability and Phlegm Syndrome and Therapeutic Potential of Some Medicinal Herbs
Previous Article in Special Issue
Direct Effects of D-Chiro-Inositol on Insulin Signaling and Glucagon Secretion of Pancreatic Alpha Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Irisin and Incretin Hormones: Similarities, Differences, and Implications in Type 2 Diabetes and Obesity

Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
*
Author to whom correspondence should be addressed.
Submission received: 1 January 2021 / Revised: 9 February 2021 / Accepted: 12 February 2021 / Published: 15 February 2021
(This article belongs to the Special Issue Pancreatic Islets of Langerhans: Not Only Beta-Cells)

Abstract

:
Incretins are gut hormones that potentiate glucose-stimulated insulin secretion (GSIS) after meals. Glucagon-like peptide-1 (GLP-1) is the most investigated incretin hormone, synthesized mainly by L cells in the lower gut tract. GLP-1 promotes β-cell function and survival and exerts beneficial effects in different organs and tissues. Irisin, a myokine released in response to a high-fat diet and exercise, enhances GSIS. Similar to GLP-1, irisin augments insulin biosynthesis and promotes accrual of β-cell functional mass. In addition, irisin and GLP-1 share comparable pleiotropic effects and activate similar intracellular pathways. The insulinotropic and extra-pancreatic effects of GLP-1 are reduced in type 2 diabetes (T2D) patients but preserved at pharmacological doses. GLP-1 receptor agonists (GLP-1RAs) are therefore among the most widely used antidiabetes drugs, also considered for their cardiovascular benefits and ability to promote weight loss. Irisin levels are lower in T2D patients, and in diabetic and/or obese animal models irisin administration improves glycemic control and promotes weight loss. Interestingly, recent evidence suggests that both GLP-1 and irisin are also synthesized within the pancreatic islets, in α- and β-cells, respectively. This review aims to describe the similarities between GLP-1 and irisin and to propose a new potential axis–involving the gut, muscle, and endocrine pancreas that controls energy homeostasis.

1. Introduction

Incretins are gut hormones that potentiate insulin secretion after meal ingestion in a glucose-dependent manner. In particular, the “incretin effect” refers to the ability of oral glucose to elicit a greater insulin secretory response than does intravenous glucose, despite inducing similar levels of glycaemia [1]. The two best-studied incretins, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), exert insulinotropic actions through distinct G-protein-coupled receptors (GIP-R and GLP-1R respectively) that are highly expressed on islet β-cells but also on α- and δ-cells, as well as in nonislet cells (e.g., heart, gastrointestinal tract, kidney, and several regions of the central nervous system [CNS]; GIP-R is also expressed in bone and adipose tissue, while the presence of GLP-1R in human adipose tissue, liver, or skeletal muscle is equivocal) [2,3]. Increasing evidence suggests that GLP-1 is also produced by the pancreatic α-cells and can therefore be considered a new pancreatic islet hormone exerting insulinotropic and glucagonostatic effects locally via paracrine and/or autocrine actions [4].
On the other hand, irisin is a myokine firstly described in 2012 by Boström et al. [5] as a peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC1-α)-dependent myokine, secreted following physical activity and able to drive brown fat-like development of white adipose tissue and thermogenesis. Many studies have explored the pleiotropic properties of irisin, demonstrating its pivotal role in the regulation of energy metabolism, by acting on several tissues and intervening in numerous biochemical pathways. Recently, it has been proposed that irisin could exert its effects via αV integrin receptors [6]. Although skeletal muscle accounts for approximatively 72% of the total amount of irisin in the circulation [5], several studies suggest that irisin can also be produced by the pancreatic islets [7,8,9], thus emerging as a new potential intra-islet hormone.
Interestingly, irisin and GLP-1 show comparable pancreatic and pleiotropic effects and activate similar intracellular pathways.
In light of this evidence, the current review aims to describe the similarities between GLP-1 and irisin and to propose a new potential axis—involving the gut, muscle, and endocrine pancreas—relevant for the control of energy homeostasis.

2. GLP-1 and Irisin Synthesis, Secretion, and Mechanism of Action: So Different but so Similar

GLP-1 is synthesized and secreted from enteroendocrine L cells scattered throughout the small and large intestine, following the post-translational processing of proglucagon by prohormone convertase 1/3 (PC1/3) (Figure 1a).
In its biologically active form, GLP-1 is a 31-amino acid peptide designated as GLP-1(7-37). This peptide can be amidated at the C-terminal glycine residue to give rise to a second form of 30-amino acid peptide, GLP-1(7–36)NH2, that is biologically equipotent to GLP-1(7–37) and represents the major form in humans [10] (Figure 1c). The constant basal secretion of GLP-1 from enteroendocrine cells is rapidly augmented by the ingestion of luminal nutrients, mainly carbohydrates and fats and, to a lesser extent, proteins [11]. In addition, GLP-1 release can be stimulated by individual nutrients, including monosaccharides (glucose, galactose and fructose), fatty acids (FAs)–especially unsaturated FAs–and amino acids [3,12]. GLP-1 secretion occurs in a biphasic pattern starting with an early (within 10–15 min) phase that is followed by a longer (30–60 min) second phase [13]. In humans, the basal concentration of bioactive GLP-1 is 5–10 pmol/L and increases approximately 2–to 3-fold after a meal, depending on both the size and nutrient composition of the meal [3]. The half-life of GLP-1 in the circulation is less than 2 min due to rapid inactivation by the ubiquitous proteolytic enzyme dipeptidyl peptidase-4 (DPP-4) [14]. The GLP-1R belongs to the class B family of seven-transmembrane-spanning, heterotrimeric G-protein-coupled receptors. GLP-1R binding GLP-1 leads to the formation of cyclic adenosine monophosphate (cAMP) and the activation of phospatidylinositol-3 kinase (PI-3K), as well as related downstream pathways [3] (Figure 2).
Irisin is a 112-amino acid hormone (~12 KDa) that results from the proteolytic cleavage of the extracellular, N-terminal portion of the membrane protein fibronectin type III domain containing protein 5 (FNDC5; Figure 1b) [5]. Although the proteases responsible for FNDC5 cleavage are not yet fully identified, Yu et al. [15] have proposed that the metalloproteinase ADAM10 (A Disintegrin And Metalloproteinase domain-containing protein 10) could be the candidate enzyme responsible for irisin release. The amino acid sequences of irisin and GLP-1 are completely dissimilar (Figure 1c). The main stimulus to irisin expression and secretion is physical activity, varying in relation to intensity, type, time, and frequency [16]. A recent meta-analysis found that irisin concentration increased by ~15% immediately following an acute bout of exercise and identified fitness level as the single best predictor—being fit was associated with a nearly two-fold increase in post-exercise irisin concentration, compared with being unfit [17]. Considerable uncertainty remains regarding the veracity of methods used for the dosage of serum irisin [18]. Although Jedrychowski et al. [19] have indisputably detected and quantified circulating human irisin by tandem mass spectrometry, assessing that its concentration is ~3.6 ng/mL (~300 pmol/L) in sedentary humans and ~4.3 ng/mL (~358 pmol/L) in individuals undergoing aerobic interval training, currently available ELISA assays for quantifying circulating irisin levels still lack of quality and accuracy, and the measured values are very dissimilar between the different assays [18,20]. This may partially explain the discrepant data found by different research groups, especially in the setting of serum irisin measurements. As previously mentioned, irisin could exert its effects via αV integrin receptors, although further investigation is required to endorse this discovery [6]. α-integrins bind β-integrins to form obligate heterodimers that, upon ligand binding, usually trigger canonical signaling via phosphorylation of focal adhesion kinase (FAK), AKT/protein kinase B (PKB), and cAMP response element-binding protein (CREB) [21,22]. Indeed, GLP-1R and integrins, although structurally very different, share numerous intracellular signaling mediators (Figure 2, green boxes).
In addition to physical activity, the nutritional composition of the diet could also impact irisin secretion. We have previously demonstrated that a high-fat diet (HFD, 60% of energy from fat) causes a rapid and persistent increase of blood irisin concentrations, in healthy, wild-type mice [23]. In particular, irisin release from myotubes could also be influenced by the type of FA, as it appears to be stimulated by saturated, but not by monounsaturated, FAs [23]. Accordingly, Guilford et al. [24] showed that FNDC5 mRNA levels were increased in skeletal muscle and adipose tissue of HFD-fed mice. It has also been demonstrated in humans that the intake of saturated FAs with diet could increase irisin release [25]. Increased FNDC5/irisin may be a compensatory mechanism to offset HFD-induced weight gain and insulin resistance by increasing energy expenditure [24]. Conversely, de Macêdo et al. [26] showed that a high-carbohydrate diet and a HFD, but not a high-protein diet, reduced irisin protein expression in the soleus muscle of female mice. Importantly, in that study, circulating irisin levels were not measured, and this could explain the discrepancy with previous literature, since it has been demonstrated that regulation of irisin expression and its cleavage by FAs are two independent events [23]. Other studies failed to detect significant changes in circulating irisin levels [27] or found reduced irisin levels [28] in rats fed an HFD for 10 or 6 weeks, respectively. In those studies, however, irisin levels were measured only at the end of the nutritional intervention, whereas changes in irisin levels could be an acute response to nutrient ingestion, rather than a long-lasting effect. In obese subjects with metabolic syndrome, irisin concentrations were equally reduced after two weight loss diets differing in macronutrient composition; however, they were positively correlated with carbohydrate intake coming from cereals, pulses, fruit, and vegetables [29]. Similarly, irisin levels were directly associated with healthy diet types and patterns, particularly with increasing fruit consumption [30]. In contrast, Park et al. demonstrated that diet quality was not associated with irisin levels in humans [31]. Although limited and confounding data exist regarding the effect of the macronutrient composition of the diet on irisin levels—possibly due to differences in animal models or human cohorts, experimental diets, and methods and timing used for irisin measurement—meal composition does appear to affect irisin secretion, as is the case with GLP-1 (Table 1).

3. Incretins and Irisin in Type 2 Diabetes and Obesity: Two Sides of the Same Coin

It has been amply demonstrated that the “incretin effect” is typically reduced or even absent in people with impaired glucose tolerance or diabetes, and this contributes to defective insulin secretion and hyperglycaemia in patients with type 2 diabetes (T2D) [1]. As no consistent differences exist in the secretion of GIP [32] or GLP-1 [33] between individuals with and without T2D, the reduction of the “incretin effect” is the result of the total loss of the insulinotropic response to GIP and the reduced ability of GLP-1 to induce insulin secretion, likely due to a reduction of functional β-cell mass and receptor activity [1]. Because GLP-1, unlike GIP, retains substantial insulinotropic activity in diabetic patients, GLP-1R agonists (GLP-1RAs) and DPP-4 inhibitors are widely used as antidiabetic therapies. GLP-1RAs are particularly appreciated not only for their antihyperglycemic action but also for their cardiovascular benefits [34], as well as their ability to promote weight loss [35] and preserve/restore β-cell functional mass [36]. To date, exenatide, lixisenatide, liraglutide, dulaglutide, and semaglutide are the available GLP-1RAs.
Conversely, in a state of obesity, GIP levels are elevated during both fasting and after an oral glucose challenge, whereas GLP-1 levels are unchanged during fasting and reduced in response to an oral glucose challenge [37]. Overall, the “incretin effect” has been reported to be decreased in obesity, likely as a result of a reduced responsiveness to GIP or reduced contribution of GLP-1 to the insulin secretory response [38].
Likewise, despite the existence of conflicting results, most clinical studies, including the meta-analyses thereof, agree that circulating irisin levels are lower in patients with T2D [20,39,40,41]. In contrast, irisin levels appear to be higher in dysmetabolic states [42], such as obesity [43,44], possibly reflecting a condition of irisin resistance or a compensatory increase for the metabolic abnormalities and insulin resistance characteristic of these patients [44]. In addition, some studies have reported positive correlations between serum irisin levels and markers of adiposity (fat mass, waist circumference, waist-to-hip ratio) and between serum irisin levels and glucose and lipid homeostasis disturbances [44,45,46,47,48,49,50,51].
Numerous interventional studies in animal models of diabetes and/or obesity have shown that the exogenous administration of recombinant irisin can restore glucose and lipid homeostasis, thus exerting antidiabetic and antiobesity effects [5,52,53,54,55,56,57]. Taken together, these studies provide evidence regarding the involvement of irisin in the pathogenesis of metabolic diseases, supporting its possible therapeutic use.
Interestingly, metformin, the current first-line antidiabetic drug, is able to promote both the secretion of GLP-1, likely through indirect mechanisms [58] and the synthesis of irisin in rat pancreatic β-cells [9]. Finally, it has been demonstrated that obese T2D patients treated with exenatide for 12 weeks show significantly increased irisin levels, and this increase is correlated with an improvement in the metabolic profile [59], thus suggesting the existence of a possible interplay between irisin and incretin hormones.

4. Effects of GLP-1 and Irisin on Function and Survival of Pancreatic Islets

A deficit of β-cell functional mass is recognized as a necessary and early condition for the development of T2D. Thus, β-cell regeneration or the preservation of functional pancreatic islet integrity should be highly considered for T2D treatment and, possibly, cure [36].
In pancreatic β-cells, in addition to their known ability to induce glucose-stimulated insulin secretion (GSIS), GLP-1R activation results in increased glucose sensitivity, insulin biosynthesis, and β-cellular neogenesis and proliferation, while decreasing apoptosis [36] (Figure 3a). Indeed, numerous studies have demonstrated that GLP-1RAs are able to protect or restore human pancreatic β-cell function and mass, both in vitro and in vivo [36]. In addition, GLP-1 also inhibits glucagon secretion from pancreatic α-cells [60,61] (Figure 3a).
GLP-1 stimulates insulin secretion by intervening in nearly all steps involved in the exocytosis of insulin secretory granules (i.e., ATP synthesis, inhibition of potassium-ATP channels [KATP], increase in intracellular Ca2+ levels) [3]. These events are mainly regulated by cAMP-protein kinase A (PKA) and cAMP-Epac2 (exchange protein directly activated by cAMP 2) signaling pathways [62] (Figure 3b). In particular, the cAMP-PKA pathway controls the inhibition of KATP, and the cAMP-Epac2 pathway endorses the synthesis of ATP, while both signaling pathways promote the increase of intracellular Ca2+ levels [62]. Importantly, GLP-1 induces insulin secretion in a glucose-dependent manner, although the mechanisms are not yet fully understood. In addition, GLP-1 also increases proinsulin gene expression and insulin biosynthesis, through a mechanism involving cAMP/PKA-dependent and -independent signaling pathways [62,63].
The proliferative effects of GLP-1 require the activation of numerous intracellular mediators, particularly PI-3K, AKT/PKB, protein kinase C (PKC), insulin receptor substrate-2 (IRS-2), mitogen-activated protein kinase (MAPK), and extracellular signal-regulated kinases (ERK), leading to the upregulation of transcription factors, such as pancreatic and duodenal homeobox 1 (PDX-1) and CREB, and of the cell cycle regulator cyclin D1, all involved in the proliferation process (Figure 3b). These mechanisms have been demonstrated mostly in rodent models, since human β-cell proliferation in vitro is rarely observed. The protective effects of GLP-1 and GLP-1RAs against various toxic stimuli, including high-glucose, FAs, cytokines, and reactive oxygen species, have also been extensively investigated and involve multiple signaling pathways, most of which overlap with those inducing cell proliferation (i.e., PI-3K, AKT/PKB, PDX-1, CREB) and lead to the upregulation of antiapoptotic proteins, such as Bcl-2 and Bcl-xl, and downregulation of proapoptotic proteins, such as Bax, Bad, and caspases [62] (Figure 3b). We have previously demonstrated, in rat and mouse β-cell lines, as well as in human and murine pancreatic islets, that exendin-4 is able to prevent TNFα- [64] and palmitic acid-induced β-cell apoptosis [65] by reducing the ability of these stressful stimuli to activate proapoptotic effectors, such as the stress kinases c-jun N-terminal kinase (JNK) [64,65] and p38 MAPK [65], and the redox adaptor protein p66Shc [66]. Finally, GLP-1 is also able to preserve β-cell identity and promote the neogenesis of new β-cells starting from non-β-cell precursors, in both rodent models and human pancreatic ducts [62,67]. (For more detailed information concerning the mechanisms by which GLP-1 promotes β-cell function, survival, and regeneration, please see references [3,12,62]).
In pancreatic islets, irisin exerts effects that are very similar to those of GLP-1 (Figure 3a). We have previously demonstrated that recombinant irisin protects human and rodent β-cells and pancreatic islets from saturated FA-induced apoptosis by enhancing AKT/Bcl-2 antiapoptotic signaling [23] (Figure 3b). However, unlike GLP-1, irisin had no effects on FA-induced JNK, p38 MAPK, or p66Shc activation (unpublished data). Furthermore, irisin promoted proinsulin mRNA transcription and increased insulin content and secretion, in a PKA/CREB-dependent manner, as the PKA inhibitor H-89 abolished all these effects [23] (Figure 3b). The exact mechanism by which irisin induces insulin secretion is not yet known, however. Moreover, similar to GLP-1, irisin induced cAMP generation [23], suggesting the existence of a specific receptor.
In addition, irisin promoted β-cell proliferation through the activation of the ERK1/2 pathway [23]. Additionally, when administered in vivo, irisin improved GSIS and increased insulin content and β-cell mass and proliferation in healthy wild type mice, while only slightly reducing α-cell mass, although glucagon secretion was not investigated [23]. Importantly, both in vivo and in vitro, irisin enhanced insulin secretion only in a glucose-stimulated manner and not under low-glucose conditions, which can minimize the risk of hypoglycemia [23]. In accordance with these results from our group, Liu et al. [57] demonstrated that irisin significantly increased the proliferation of rat insulin secreting INS-1 cells via the ERK and p38 MAPK signaling pathways, protected the cells from high glucose-induced apoptosis by regulating the expression of pro- (Bax, Bad and caspases) and antiapoptotic (Bcl-2 and Bcl-xl) proteins, and improved pancreatic β-cell function in a T2D model in male rats. Furthermore, Zhang et al. [68] showed that irisin enhanced the expression of genes related to β-cell survival (e.g., PDX-1/Bcl-2) and function (e.g., GLUT2/glucokinase), thus reversing glucolipotoxicity-induced apoptosis and restoring insulin secretory ability in β-cells under glucolipotoxic conditions (Figure 3b). In particular, these effects were dependent on the activation of adenosine monophosphate-activated protein kinase (AMPK), the suppression of lipogenic gene expression (e.g., acetyl-CoA carboxylase), and therefore the reduced synthesis and intracellular accumulation of FAs/triglyceride [68]. Irisin also decreased the expression of proinflammatory genes [68].
Recently, it has been suggested that β-cells themselves could produce irisin, which could enhance β-cell survival and function by acting in autocrine-manner [9].

5. Extra-Pancreatic Effects of GLP-1 and Irisin: Beyond Glycemic Control

The advent of GLP-1RAs represented a revolution in the treatment of T2D, because in addition to ensuring good glycemic control with a low risk of hypoglycemia and weight loss, they showed cardiovascular protective effects. These effects are in part dependent on the reduction of hyperglycemia but also involve direct effects of GLP-1 on several organs and tissues, such as the heart and vessels, CNS, and, to a lesser extent, kidney, liver, skeletal muscle, and adipose tissue. Likewise, irisin has shown similar pleiotropic effects (Figure 4).

5.1. Cardiovascular System

Although best known for their insulinotropic and weight-lowering actions, GLP-1 and GLP-1RAs also confer a series of beneficial effects on the cardiovascular system. These include the inhibition of cardiomyocyte apoptosis induced by different harmful stimuli [69,70,71,72,73,74], amelioration of endothelial dysfunction [75,76,77], and improvement of myocardial function and cardiac output in experimental models of cardiac injury or heart failure [69,78,79,80]. Indeed, GLP-1 reduces infarct size in the isolated perfused rat heart and in animal models of myocardial ischemia [81,82]. In addition, while some studies report increased heart rate (HR) and blood pressure (BP) in rodents upon acute or chronic GLP-1R agonism [83,84,85], other reports show decreased BP, especially in experimental models associated with the development of hypertension [86,87]. In humans, GLP-1RAs have shown moderate stimulatory effects on HR with reduced systolic BP in hypertensive and T2D individuals [88,89,90,91], and these effects are likely obtained through vasodilation and activation of nitric oxide-dependent mechanisms [12,91]. GLP-1RAs have also been demonstrated to exert a proangiogenic and antiapoptotic effect in human endothelial cells in vitro [92,93,94]. In addition, they have shown an antiatherogenic property both in animal models [95,96,97,98] and humans [99] (Figure 4). Finally, we have previously demonstrated that GLP-1RAs are also able to prevent palmitic acid-induced apoptosis [100] and H2O2-induced oxidative stress in human cardiac progenitor cells [101], which are essential for constant tissue repair and renewal in the adult heart.
Similarly, several recent reports have shown that irisin protects the heart against ischemia/reperfusion injury, improves cardiac function, and reduces infarct size, both in diabetic and nondiabetic rodents [102,103,104]. In addition, irisin improves myocardial performance (increased ejection fraction and reduced fibrosis) in db/db mice characterized by depression in cardiac function [105]. Interestingly, in a mouse model of acute myocardial infarction (MI), irisin administration significantly reduced infarct size and fibrosis, and alleviated MI-induced cardiac dysfunction and ventricular dilation [106]. Importantly, irisin administration significantly increased angiogenesis in the infarct border zone and decreased cardiomyocyte apoptosis but did not influence cardiomyocyte proliferation [106]. Accordingly, irisin has been shown to protect cardiomyocytes against apoptosis and dysfunction induced by different stressful stimuli, such as lipotoxicity [107], H2O2 [108], hypoxia-reoxygenation [109,110], and doxorubicin-induced oxidative stress [111], as well as alleviating angiotensin-II-induced cardiac hypertrophy and fibrosis [15,112,113] and pressure overload-induced cardiac hypertrophy [114]. The induction of autophagy has been proposed as a mechanism though which irisin exerts these beneficial effects [112,114,115], as block of the autophagic flux has been identified as a possible cause of cardiomyocyte apoptosis and cardiac hypertrophy. In addition, the activation of AKT [108,111,116] and AMPK [15,109,114] could represent key signaling steps in the cardio-protective effects of irisin. Moreover, irisin could also promote cardiac progenitor cell-induced myocardial repair and functional improvement in an infarcted heart [117]. Additionally, irisin was able to favor endothelium-dependent vasodilation and therefore to lower BP in obese mice and in the spontaneously hypertensive rat, through an AMPK-AKT-endothelial nitric oxide synthase (eNOS) dependent mechanism [118,119,120,121]. The BP-lowering effect of irisin has been examined in several studies. Peripheral administration of irisin reduced BP in both control and spontaneously hypertensive rats [122,123] through several different mechanisms, including induction of mesenteric arteries relaxation [122,124], activation of neurons controlling cardiac vagal tone [125], and augmentation of acetylcholine-mediated vasodilation [121].
Furthermore, in vitro studies conducted on human umbilical vein endothelial cells (HUVECs) or human microvascular endothelial cells (HMEC-1) have shown the irisin’s ability to exert proangiogenic effects through activation of the ERK-regulated proliferation signaling pathway [106,126,127,128], to prevent apoptosis induced by high fat, high glucose, or advanced glycation end products [128,129,130] and to counteract the impairment of angiogenesis by oxidized low-density lipoprotein (LDL) [116]. Interestingly, irisin could also improve endothelial function by increasing the number of endothelial progenitor cells (EPCs) in peripheral blood of diabetic mice and by improving the function of bone marrow-derived EPCs in diabetic mice via the PI3K-AKT-eNOS pathway [131]. Finally, irisin is able to inhibit atherosclerosis in apolipoprotein E-deficient mice by suppressing oxidized LDL-induced vascular inflammation and endothelial dysfunction [132] and promoting endothelial cell proliferation [127], likely through the AMPK-PI3K-AKT-eNOS signaling pathway [133] (Figure 4).

5.2. Central Nervous System

Both central and peripheral administration of GLP-1 and its analogs are able to lower body weight via centrally regulated inhibition of food intake in both healthy individuals and patients with T2D [134,135,136,137,138]. Of note, in rodents, intravenous infusion of liraglutide inhibits eating by directly promoting the expression of the hypothalamic anorexigenic genes cocaine- and amphetamine-regulated transcript (CART) and pro-opiomelanocortin (POMC) in the arcuate nucleus [139]. Importantly, liraglutide failed to suppress food intake in CNS-specific GLP-1R KO mice [140]. Consistent with this, in rats, central administration of the GLP-1R inhibitor exendin (9–39) was sufficient to attenuate the anorexigenic effect of peripherally administered liraglutide and exendin-4 [141], suggesting the importance of GLP-1R activation in the brain, and specifically in hypothalamic neurons, for the anorexigenic effects of GLP-1 and its analogs (Figure 4). Importantly, although GLP-1 and GLP-1RAs of relatively small molecular size (liraglutide, lixisenatide, and exendin-4) have all been demonstrated to cross the blood brain barrier (BBB) upon peripheral administration [142,143], the hypothalamus and hindbrain—both areas with incomplete BBB—appear to be the main targets of peripherally injected GLP-1RAs [12]. However, GLP-1RAs can also act on vagal afferents to transmit the signal to the hindbrain, which then projects to other key feeding areas in the hypothalamus [12]. Interestingly, GLP-1 is also synthesized by the hindbrain in a discrete set of neurons within the nucleus of the solitary tract [144,145,146], but little is known regarding the neurophysiology of these neurons and whether it is peripheral or central secretion of GLP-1 that regulates food intake.
On the other hand, the ability of irisin to modulate food intake in diabetic or obese mice/rats remains little investigated and the few existing results are conflicting [53,54]. We have recently demonstrated that daily intraperitoneal injection of irisin for 14 days in healthy C57BL/6 mice increases mRNA levels of the hypothalamic anorexigenic genes CART and POMC, similar to GLP-1, without affecting mRNA expression of the orexigenic genes agouti-related neuropeptide (AgRP), orexin, and promelanin-concentrating hormone (PMCH), and without inducing apparent changes in food intake [147]. Similarly, Butt et al. [148] showed that intraperitoneal irisin injection in goldfish increased expression of CART mRNA in the brain, without affecting AgRP mRNA expression, and induced a decrease in food intake, confirming that irisin may act as an anorexigenic factor in fish. In another study, intrahypothalamic injection of irisin for 24 h in rats augmented CART/POMC and reduced orexin mRNA levels, without affecting mRNA levels of the orexigenic neuropeptide Y (NPY) and AgRP, promoting a reduction in food intake [149]. Conversely, in another study in nonobese, nondiabetic rats, intracerebroventricular infusion of irisin for 7 days increased food intake and was associated with decreased POMC mRNA levels in the hypothalamus [150]. Considering these results, it is possible that the effects of irisin administration on food intake may be dependent upon the timing and/or route of administration (Figure 4). Importantly, like GLP-1, irisin can be locally expressed in the brain, since this peptide has been detected in the cerebrospinal fluid, and FNDC5 is known to be highly expressed in the glia (e.g., astrocytes and microglia) and neurons of various brain regions (e.g., cerebellar Purkinje cells, hypothalamus, hippocampus) [151,152]. Furthermore, peripheral irisin can also cross the BBB, although the mechanism of transport from the blood to the cerebrospinal fluid remains unknown [153,154].
In addition to affecting feeding behavior, central GLP-1R agonism has been demonstrated to have neuroprotective effects [155,156] and to improve several aspects of learning and memory [157]. The molecular mechanisms underlying the neuroprotective effects of central GLP-1R agonism are, at least in part, mediated by cAMP formation and subsequent PI-3K and ERK activation [12]. Impaired cerebral glucose metabolism and brain insulin resistance are main pathological features of Alzheimer disease (AD), and this has recently led researchers to designate AD as “diabetes of the brain” or “type 3 diabetes” [158]. Accordingly, GLP-1RAs have been tested and have shown encouraging protective actions in experimental models of AD, as well as in initial clinical trials [159,160,161,162] (Figure 4).
Similarly, several studies have demonstrated that irisin plays a developmental role in regulating the process of neuronal differentiation and maturation [152], induces the expression of neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) [147,152], and could exert neuroprotective effects on neurodegenerative diseases, improving memory impairment and synaptic plasticity [152]. Interestingly, irisin improves learning and memory function, promotes neurogenesis, and prevents the neuronal damage caused by oxidative stress, thus representing a potential future target for ameliorating AD pathology and preventing AD onset [151,163] (Figure 4).

5.3. Kidney, Liver, Skeletal Muscle, Adipose Tissue, and Bone

The GLP-1R is expressed in the kidney and intravenous infusion of GLP-1 in animal models produces natriuretic and diuretic responses that are associated with increased glomerular filtration rate and inhibition of sodium reabsorption in the proximal tubule, thus exerting an antihypertensive effect [3]. The few studies in the literature that have examined the effects of irisin on the kidneys agree that this myokine is able to improve kidney dysfunction, as well as to reduce kidney injury and fibrosis caused by various stressful stimuli [164,165,166,167,168] (Figure 4).
It is not definitively clear if the effects of GLP-1 on liver, skeletal muscle, and adipose tissue depend on a direct effect or are the result of the overall amelioration in whole-body insulin sensitivity. Indeed, the presence of GLP-1R in these tissues is equivocal [2,3,12], although more recent studies have clearly identified that GLP-1R is expressed both in skeletal muscle [169,170] and adipocytes [12]. Accordingly, in skeletal muscle, GLP-1 increases glucose uptake, insulin-stimulated glucose metabolism, and glycogen synthesis [3]. In addition, GLP-1 also enhances glucose uptake and insulin-stimulated glucose metabolism in adipocytes [3]. Moreover, in human adipocytes, GLP-1 displays both lipolytic and lipogenic actions [171], as well as adipogenic effects [172]. Finally, it has been shown that GLP-1 inhibits hepatic glucose production and increases glycogen synthesis in hepatocytes [3], whereas exendin-4 improves insulin sensitivity and reverses hepatic steatosis in ob/ob mice [173,174]. Other studies, however, do not support a direct role for GLP-1R signaling in these tissues [175,176], and whether or not GLP-1R agonists can influence glucose disposal and insulin sensitivity independent of changes in insulin or glucagon remains unclear (Figure 4).
Conversely, numerous studies have confirmed the ability of irisin to directly act on liver, skeletal muscle, and adipose tissue. Indeed, irisin has been demonstrated to inhibit lipogenesis, cholesterol synthesis, and gluconeogenesis [55,177,178,179,180,181,182,183], activate FAs oxidation and glycogen synthesis [55,181,182,183], and reduce lipid accumulation, steatosis, and insulin resistance [178,180,181] in the liver. Finally, it has been shown that irisin is able to protect hepatocytes from injury caused by different harmful stimuli, such as methotrexate [184], ischemia-reperfusion [185,186], and glucose/lipid overload [183]. These effects suggest a lipid-lowering and hepatoprotective attitude of irisin. In addition, several papers have shown that irisin can act in skeletal muscle, promoting myogenesis, muscle hypertrophy [187] and metabolism, enhancing insulin sensitivity under basal [188] or lipotoxic [189,190,191] conditions, and protecting myocytes from high glucose- and FAs-induced cytotoxicity [188]. In particular, irisin improves glucose uptake [55,188,192,193,194] and inhibits gluconeogenesis and glycogenolysis [194], while increasing glycogen storage [188,194] and stimulating FAs oxidation [55,194,195]. The activation of AMPK appears to be a key signaling step for irisin action in the liver and skeletal muscle. In adipose tissue, it has been shown that irisin reduces adipogenic differentiation of preadipocytes [196,197] and promotes glucose uptake and glucose and lipid metabolism in mature adipocytes [197,198]. In addition, overexpression of FNDC5 in obese mice reduces the size of adipocytes in subcutaneous adipose tissue and stimulates lipolysis [53]. Adipocytes isolated from animals injected with irisin, or treated ex vivo, are therefore smaller and accumulate fewer lipids than do controls [53]. Furthermore, irisin treatment reduces lipid accumulation in human and mouse adipocytes [198,199] (Figure 4). According to Boström et al. [5], irisin also drives the browning of white adipose tissue by stimulating the expression of uncoupling protein-1 (UCP-1) and other known brown fat genes, while downregulating genes characteristic of white fat development, thus promoting thermogenesis and energy expenditure. In human adipocytes, the ability of irisin to induce browning is still debated: it is likely that only a small subpopulation of adipocytes that highly express “brite”-specific markers is responsible for the irisin effect [200]. In addition, Zhang et al. [196] suggested that the effects of irisin in humans are likely differentiation stage-dependent, as irisin induces browning in mature adipocytes but not in preadipocytes (Figure 4).
It has been reported that GLP-1 can promote bone formation and inhibit bone resorption in animal models, thus enhancing bone mineral density and quality [201] (Figure 4). These effects are of interest for T2D patients, as they are characterized by higher risk of bone fractures compared with healthy individuals [202], likely due to a reduction in bone turnover [203]. Promotion of GLP-1-induced bone formation may depend on the increase in the number of osteoblasts [204], in the expression of genes [205,206] and serum markers [205,207] related to bone formation, as well as, indirectly, on the reduction of glycemia [201]. All these effects are possibly mediated by MAPK [208] and Wnt [209,210] pathways, or c-fos transcription promotion [201]. On the other hand, GLP-1 inhibits bone resorption probably by reducing the number of osteoclasts [205] and promoting calcitonin secretion [201]. Unfortunately, the specific process and related molecular pathways by which GLP-1 acts on the bone are still not completely understood, and the relationship between GLP-1 and bone fractures is still under investigation [201]. Interestingly, irisin has also shown to exert anabolic effects on bone in mice [211]. Specifically, irisin promotes osteoblast differentiation [212], increases cortical bone mass, and makes bone more resistant and less susceptible to fractures [213] (Figure 4). As for GLP-1, also irisin effects could depend upon the MAPK pathway activation [214]. Additionally, in humans, irisin was inversely correlated with the incidence of bone fractures in postmenopausal osteoporotic women, as well as in patients with T2D, cardiovascular disease, and liver disease [215].

6. GIP and Irisin: Similarities and Differences Not to Be Underestimated

In this review we have focused on the similarities between irisin and GLP-1 because of the therapeutic potential of GLP-1, rather than GIP, in T2D. Nevertheless, there are similarities and differences also between irisin and GIP that should not be underestimated.
GIP is released by intestinal K cells and, similar to GLP-1, is secreted in response to nutrient ingestion and activates GIP-R in pancreatic β-cells, enhancing meal-stimulated insulin secretion in a glucose-dependent manner and stimulating β-cell survival and, especially in rodents, proliferation [3]. In addition, GIP has similar effects to GLP-1 in the hypothalamic regulation of food intake [38,216]. Because improving glucose control with antidiabetic therapies appears to restore the insulinotropic effect of GIP in patients with T2D, growing evidence suggests that GLP-1R/GIP-R co-agonists could represent a promising strategy for treating T2D and obesity [217]. In particular, the overlapping of insulinotropic and body weight-lowering actions of both GLP-1 and GIP may have the potential to bolster their glucose-lowering and appetite-suppressing effects beyond what is observed with individual agents [217]. Importantly, GIP also targets bone similar to irisin, inhibiting bone resorption while promoting its formation, and thus exerting anabolic effects [3,218,219] (Figure 5).
Moreover, GIP also targets adipose tissue, stimulating adipogenesis, lipogenesis, and lipid storage in fat cells [3,217]. It has been suggested that the increase in lipid-buffering capacity of adipose tissue reduces lipid “spillover” and ectopic fat accumulation in tissues such as liver, skeletal muscle, heart, and pancreas, thus improving insulin sensitivity [220]. In contrast, as mentioned in the previous paragraph, the overall effects of irisin on adipose tissue lead to a reduction in fat mass and an increase in energy expenditure (Figure 5), which together may promote weight loss.

7. Conclusions

By definition, incretins are hormones with low basal plasma concentrations that are released after the ingestion of physiological nutrients to reach concentrations that augment the insulin secretory responses at a permissive degree of hyperglycaemia, although they are ineffective at low glucose concentrations [221]. According to this definition, irisin can be considered an incretin-like hormone, as its secretion appears to be also affected by meals and, once secreted, it enhances insulin secretion in a glucose-dependent manner. In addition, both the GLP-1 insulinotropic effects and irisin levels are defective in T2D, and their exogenous administration is able to improve glycemic control. Importantly, irisin and GLP-1 share numerous direct pancreatic effects (Figure 3), including the ability to induce insulin biosynthesis and glucose-dependent insulin secretion, as well as improving β-cell proliferation and survival. Albeit through different pathways, irisin and GLP-1 activate the same intracellular signaling proteins (e.g., AKT, CREB, ERK1/2; Figure 3b). Interestingly, irisin shows cardiovascular and anorexigenic effects similar to those of GLP-1, while demonstrating a more pronounced beneficial metabolic effect in insulin-dependent organs, such as liver, skeletal muscle, and adipose tissue (Figure 4). Of note, because GLP-1 and irisin signal through different receptors that nevertheless converge on very similar pathways (Figure 2 and Figure 3b), an additive effect of the two molecules cannot be excluded.
Additionally, like GIP, irisin promotes bone formation while, unlike GIP, it reduces lipid accumulation in adipose tissue (Figure 4 and Figure 5). Irisin could therefore broaden the beneficial effects of GLP-1RAs by restoring bone turnover and improving weight loss through the reduction of fat mass. Furthermore, recent evidence suggests that both GLP-1 and irisin are also synthesized within the pancreatic islets, lengthening the list of intra-islet hormones and paving the way for new scenarios in interorgan cross-talk.
In conclusion, due to its modalities of secretion and its pancreatic and extra-pancreatic effects, irisin could be considered an incretin-like hormone with an action similar to that of GLP-1, with the addition of an anabolic GIP-like effect at the bone level and a marked antilipogenic action. This evidence highlights the promising antidiabetic and antiobesity potential of irisin, possibly in combination therapeutic regimens with GLP-1RAs.

Author Contributions

All authors contributed to the conception, literature search, figure design and writing of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by European Union–European Social Fund, PON R&I 2014-2020, AIM 1810057, and the “Fondazione per la Ricerca Biomedica Saverio e Isabella Cianciola”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

A.N., N.M. and F.G. are named inventors of a patent related to the work described. G.P., A.B., A.C., S.P., and L.L. declare no conflict of interest.

References

  1. Nauck, M.A.; Meier, J.J. The incretin effect in healthy individuals and those with type 2 diabetes: Physiology, pathophysiology, and response to therapeutic interventions. Lancet Diabetes Endocrinol. 2016, 4, 525–536. [Google Scholar] [CrossRef]
  2. 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]
  3. Baggio, L.L.; Drucker, D.J. Biology of Incretins: GLP-1 and GIP. Gastroenterology 2007, 132, 2131–2157. [Google Scholar] [CrossRef]
  4. Fava, G.E.; Dong, E.W.; Wu, H. Intra-islet glucagon-like peptide 1. J. Diabetes Complicat. 2016, 30, 1651–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Boström, P.; Wu, J.; Jedrychowski, M.P.; Korde, A.; Ye, L.; Lo, J.C.; Rasbach, K.A.; Bostroem, E.A.; Choi, J.H.; Long, J.Z.; et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nat. Cell Biol. 2012, 481, 463–468. [Google Scholar] [CrossRef] [PubMed]
  6. Kim, H.; Wrann, C.D.; Jedrychowski, M.; Vidoni, S.; Kitase, Y.; Nagano, K.; Zhou, C.; Chou, J.; Parkman, V.-J.A.; Novick, S.J.; et al. Irisin Mediates Effects on Bone and Fat via αV Integrin Receptors. Cell 2018, 175, 1756–1768.e17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Aydin, S.; Kuloglu, T.; Aydin, S.; Kalayci, M.; Yilmaz, M.; Cakmak, T.; Albayrak, S.; Gungor, S.; Colakoglu, N.; Ozercan, I.H. A comprehensive immunohistochemical examination of the distribution of the fat-burning protein irisin in biological tissues. Peptides 2014, 61, 130–136. [Google Scholar] [CrossRef]
  8. Aydin, S. Three new players in energy regulation: Preptin, adropin and irisin. Peptides 2014, 56, 94–110. [Google Scholar] [CrossRef]
  9. Li, Q.; Jia, S.; Xu, L.; Li, B.; Chen, N. Metformin-induced autophagy and irisin improves INS-1 cell function and survival in high-glucose environment via AMPK/SIRT1/PGC-1α signal pathway. Food Sci. Nutr. 2019, 7, 1695–1703. [Google Scholar] [CrossRef] [Green Version]
  10. Opinto, G.; Natalicchio, A.; Marchetti, P. Physiology of incretins and loss of incretin effect in type 2 diabetes and obesity. Arch. Physiol. Biochem. 2013, 119, 170–178. [Google Scholar] [CrossRef]
  11. Diakogiannaki, E.; Gribble, F.M.; Reimann, F. Nutrient detection by incretin hormone secreting cells. Physiol. Behav. 2012, 106, 387–393. [Google Scholar] [CrossRef] [Green Version]
  12. Müller, T.; Finan, B.; Bloom, S.; D’Alessio, D.; Drucker, D.; Flatt, P.; Fritsche, A.; Gribble, F.; Grill, H.; Habener, J.; et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef]
  13. Herrmann, C.; Göke, R.; Richter, G.; Fehmann, H.-C.; Arnold, R.; Göke, B. Glucagon-Like Peptide-1 and Glucose-Dependent Insulin-Releasing Polypeptide Plasma Levels in Response to Nutrients. Digestion 1995, 56, 117–126. [Google Scholar] [CrossRef]
  14. Deacon, C.F.; Nauck, M.A.; Toft-Nielsen, M.; Pridal, L.; Willms, B.; Holst, J.J. Both Subcutaneously and Intravenously Administered Glucagon-Like Peptide I Are Rapidly Degraded from the NH2-Terminus in Type II Diabetic Patients and in Healthy Subjects. Diabetes 1995, 44, 1126–1131. [Google Scholar] [CrossRef] [PubMed]
  15. Yu, Q.; Kou, W.; Xu, X.; Zhou, S.; Luan, P.; Xu, X.; Li, H.; Zhuang, J.; Wang, J.; Zhao, Y.; et al. FNDC5/Irisin inhibits pathological cardiac hypertrophy. Clin. Sci. 2019, 133, 611–627. [Google Scholar] [CrossRef]
  16. Yang, D.; Yang, Y.; Li, Y.; Han, R. Physical Exercise as Therapy for Type 2 Diabetes Mellitus: From Mechanism to Orientation. Ann. Nutr. Metab. 2019, 74, 313–321. [Google Scholar] [CrossRef]
  17. Fox, J.; Rioux, B.V.; Goulet, E.D.; Johanssen, N.M.; Swift, D.L.; Bouchard, D.R.; Loewen, H.; Sénéchal, M. Effect of an acute exercise bout on immediate post-exercise irisin concentration in adults: A meta-analysis. Scand. J. Med. Sci. Sports 2017, 28, 16–28. [Google Scholar] [CrossRef]
  18. Cooke, A.B.; Gomez, Y.-H.; Daskalopoulou, S.S. 5 years later: Irisin detection still an issue. Eur. J. Endocrinol. 2017, 177, C1–C4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Jedrychowski, M.P.; Wrann, C.D.; Paulo, J.A.; Gerber, K.K.; Szpyt, J.; Robinson, M.M.; Nair, K.S.; Gygi, S.P.; Spiegelman, B.M. Detection and Quantitation of Circulating Human Irisin by Tandem Mass Spectrometry. Cell Metab. 2015, 22, 734–740. [Google Scholar] [CrossRef] [Green Version]
  20. Perakakis, N.; Triantafyllou, G.A.; Fernández-Real, J.M.; Huh, J.Y.; Park, K.H.; Seufert, J.; Mantzoros, C.S. Physiology and role of irisin in glucose homeostasis. Nat. Rev. Endocrinol. 2017, 13, 324–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Augenlicht, L.H.; Davis, R.J.; Lisanti, M.P.; Zhurinsky, J.; Troussard, A.A.; D’Amico, M.; Donehower, L.A.; Takemaru, K.-I.; Moon, R.T.; Ben-Ze’Ev, A.; et al. The Integrin-linked Kinase Regulates the Cyclin D1 Gene through Glycogen Synthase Kinase 3β and cAMP-responsive Element-binding Protein-dependent Pathways. J. Biol. Chem. 2000, 275, 32649–32657. [Google Scholar] [CrossRef] [Green Version]
  22. Giancotti, F.G. Integrin Signaling. Science 1999, 285, 1028–1033. [Google Scholar] [CrossRef] [PubMed]
  23. Natalicchio, A.; Marrano, N.; Biondi, G.; Spagnuolo, R.; Labarbuta, R.; Porreca, I.; Cignarelli, A.; Bugliani, M.; Marchetti, P.; Perrini, S.; et al. The Myokine Irisin Is Released in Response to Saturated Fatty Acids and Promotes Pancreatic β-Cell Survival and Insulin Secretion. Diabetes 2017, 66, 2849–2856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Guilford, B.L.; Parson, J.C.; Grote, C.W.; Vick, S.N.; Ryals, J.M.; Wright, D.E. Increased FNDC5 is associated with insulin resistance in high fat-fed mice. Physiol. Rep. 2017, 5, e13319. [Google Scholar] [CrossRef] [PubMed]
  25. Osella, A.R.; Colaianni, G.; Correale, M.; Pesole, P.L.; Bruno, I.; Buongiorno, C.; Deflorio, V.; Leone, C.M.; Colucci, S.C.; Grano, M.; et al. Irisin Serum Levels in Metabolic Syndrome Patients Treated with Three Different Diets: A Post-Hoc Analysis from a Randomized Controlled Clinical Trial. Nutrients 2018, 10, 844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. De Macêdo, S.M.; Lelis, D.D.F.; Mendes, K.L.; Fraga, C.A.D.C.; Brandi, I.V.; Feltenberger, J.D.; Farias, L.C.; Guimarães, A.L.S.; de Paula, A.M.B.; Santos, S.H.D.S. Effects of Dietary Macronutrient Composition on FNDC5 and Irisin in Mice Skeletal Muscle. Metab. Syndr. Relat. Disord. 2017, 15, 161–169. [Google Scholar] [CrossRef] [PubMed]
  27. Quiñones, M.; Folgueira, C.; Sanchez-Rebordelo, E.; Al-Massadi, O. Circulating Irisin Levels Are Not Regulated by Nutritional Status, Obesity, or Leptin Levels in Rodents. Mediat. Inflamm. 2015, 2015, 620919. [Google Scholar] [CrossRef] [Green Version]
  28. Lanzi, C.R.; Perdicaro, D.J.; Tudela, J.G.; Muscia, V.; Fontana, A.R.; Oteiza, P.I.; Prieto, M.A.V. Grape pomace extract supplementation activates FNDC5/irisin in muscle and promotes white adipose browning in rats fed a high-fat diet. Food Funct. 2020, 11, 1537–1546. [Google Scholar] [CrossRef]
  29. Lopezlegarrea, P.; de la Iglesia, R.; Crujeiras, A.; Casanueva, F.; Felipe, F.; Zulet, M.; Martinez, J.; José, A. Higher baseline irisin concentrations are associated with greater reductions in glycemia and insulinemia after weight loss in obese subjects. Nutr. Diabetes 2014, 4, e110. [Google Scholar] [CrossRef]
  30. Ko, B.-J.; Park, K.H.; Shin, S.; Zaichenko, L.; Davis, C.R.; Crowell, J.A.; Joung, H.; Mantzoros, C.S. Diet quality and diet patterns in relation to circulating cardiometabolic biomarkers. Clin. Nutr. 2016, 35, 484–490. [Google Scholar] [CrossRef]
  31. Park, K.H.; Zaichenko, L.; Peter, P.; Davis, C.R.; Crowell, J.A.; Mantzoros, C.S. Diet quality is associated with circulating C-reactive protein but not irisin levels in humans. Metabolism 2014, 63, 233–241. [Google Scholar] [CrossRef] [Green Version]
  32. Calanna, S.; Christensen, M.; Holst, J.J.; Laferrère, B.; Gluud, L.L.; Vilsbøll, T.; Knop, F.K. Secretion of Glucose-Dependent Insulinotropic Polypeptide in Patients with Type 2 Diabetes: Systematic review and meta-analysis of clinical studies. Diabetes Care 2013, 36, 3346–3352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Calanna, S.; Christensen, M.; Holst, J.J.; Laferrère, B.; Gluud, L.L.; Vilsbøll, T.; Knop, F.K. Secretion of glucagon-like peptide-1 in patients with type 2 diabetes mellitus: Systematic review and meta-analyses of clinical studies. Diabetology 2013, 56, 965–972. [Google Scholar] [CrossRef] [Green Version]
  34. Caruso, I.; Cignarelli, A.; Giorgino, F. Heterogeneity and Similarities in GLP-1 Receptor Agonist Cardiovascular Outcomes Trials. Trends Endocrinol. Metab. 2019, 30, 578–589. [Google Scholar] [CrossRef] [PubMed]
  35. Brown, E.; Cuthbertson, D.J.; Wilding, J.P. Newer GLP-1 receptor agonists and obesity-diabetes. Peptides 2018, 100, 61–67. [Google Scholar] [CrossRef]
  36. Marrano, N.; Biondi, G.; Cignarelli, A.; Perrini, S.; Laviola, L.; Giorgino, F.; Natalicchio, A. Functional loss of pancreatic islets in type 2 diabetes: How can we halt it? Metabolism 2020, 110, 154304. [Google Scholar] [CrossRef]
  37. Chia, C.W.; Egan, J.M. Incretins in obesity and diabetes. Ann. N. Y. Acad. Sci. 2019, 1461, 104–126. [Google Scholar] [CrossRef]
  38. Nauck, M.A.; Meier, J.J. Incretin hormones: Their role in health and disease. Diabetes Obes. Metab. 2018, 20, 5–21. [Google Scholar] [CrossRef]
  39. Munoz, I.Y.M.; Romero, E.D.S.C.; Garcia, J.D.J.G. Irisin a Novel Metabolic Biomarker: Present Knowledge and Future Directions. Int. J. Endocrinol. 2018, 2018, 1–8. [Google Scholar] [CrossRef] [Green Version]
  40. Zhang, C.; Ding, Z.; Lv, G.; Li, J.; Zhou, P.; Zhang, J. Lower irisin level in patients with type 2 diabetes mellitus: A case-control study and meta-analysis. J. Diabetes 2015, 8, 56–62. [Google Scholar] [CrossRef]
  41. Du, X.-L.; Jiang, W.-X.; Lv, Z.-T. Lower Circulating Irisin Level in Patients with Diabetes Mellitus: A Systematic Review and Meta-Analysis. Horm. Metab. Res. 2016, 48, 644–652. [Google Scholar] [CrossRef]
  42. Qiu, S.; Cai, X.; Yin, H.; Zügel, M.; Sun, Z.; Steinacker, J.M.; Schumann, U. Association between circulating irisin and insulin resistance in non-diabetic adults: A meta-analysis. Metabolism 2016, 65, 825–834. [Google Scholar] [CrossRef] [Green Version]
  43. Cao, R.Y.; Zheng, H.; Redfearn, D.; Yang, J. FNDC5: A novel player in metabolism and metabolic syndrome. Biochimie 2019, 158, 111–116. [Google Scholar] [CrossRef]
  44. Arhire, L.I.; Mihalache, L.; Covasa, M. Irisin: A Hope in Understanding and Managing Obesity and Metabolic Syndrome. Front. Endocrinol. 2019, 10, 524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Park, K.H.; Zaichenko, L.; Brinkoetter, M.; Thakkar, B.; Sahin-Efe, A.; Joung, K.E.; Tsoukas, M.A.; Geladari, E.V.; Huh, J.Y.; Dincer, F.; et al. Circulating Irisin in Relation to Insulin Resistance and the Metabolic Syndrome. J. Clin. Endocrinol. Metab. 2013, 98, 4899–4907. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, J.-J.; Wong, M.D.; Toy, W.C.; Tan, C.S.; Liu, S.; Ng, X.W.; Tavintharan, S.; Sum, C.F.; Lim, S.C. Lower circulating irisin is associated with type 2 diabetes mellitus. J. Diabetes Complicat. 2013, 27, 365–369. [Google Scholar] [CrossRef] [PubMed]
  47. Crujeiras, A.; Ana, B.; Zulet, M.; Lopez-Legarrea, P.; de la Iglesia, R.; Pardo, M.; Carreira, M.; Marcos, C.; Martinez, J.; José, A.; et al. Association between circulating irisin levels and the promotion of insulin resistance during the weight maintenance period after a dietary weight-lowering program in obese patients. Metabolism 2014, 63, 520–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Stengel, A.; Hofmann, T.; Goebel-Stengel, M.; Elbelt, U.; Kobelt, P.; Klapp, B.F. Circulating levels of irisin in patients with anorexia nervosa and different stages of obesity—Correlation with body mass index. Peptides 2013, 39, 125–130. [Google Scholar] [CrossRef] [PubMed]
  49. Huh, J.Y.; Panagiotou, G.; Mougios, V.; Brinkoetter, M.; Vamvini, M.T.; Schneider, B.E.; Mantzoros, C.S. FNDC5 and irisin in humans: I. Predictors of circulating concentrations in serum and plasma and II. mRNA expression and circulating concentrations in response to weight loss and exercise. Metabolism 2012, 61, 1725–1738. [Google Scholar] [CrossRef] [Green Version]
  50. Campolo, J.; Corradi, E.; Rizzardi, A.; Parolini, M.; Dellanoce, C.; di Guglielmo, M.L.; Tarlarini, P.; Cattaneo, M.; Trivella, M.G.; de Maria, R. Irisin and markers of metabolic derangement in non-diabetic Caucasian subjects with stage I-II obesity during early aging. PLoS ONE 2020, 15, e0229152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Saber, G.Y.; Kasabri, V.; Saleh, M.I.; Suyagh, M.; Halaseh, L.; Jaber, R.; Abu-Hassan, H.; Alalawi, S. Increased irisin versus reduced fibroblast growth factor1 (FGF1) in relation to adiposity, atherogenicity and hematological indices in metabolic syndrome patients with and without prediabetes. Horm. Mol. Biol. Clin. Investig. 2019, 38. [Google Scholar] [CrossRef]
  52. Ozgor, B.Y.; Demiral, I.; Zeybek, U.; Celik, F.; Buyru, F.; Yeh, J.; Bastu, E. Effects of Irisin Compared with Exercise on Specific Metabolic and Obesity Parameters in Female Mice with Obesity. Metab. Syndr. Relat. Disord. 2020, 18, 141–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Xiong, X.-Q.; Chen, D.; Sun, H.-J.; Ding, L.; Wang, J.-J.; Chen, Q.; Li, Y.-H.; Zhou, Y.-B.; Han, Y.; Zhang, F.; et al. FNDC5 overexpression and irisin ameliorate glucose/lipid metabolic derangements and enhance lipolysis in obesity. Biochim. Biophys. Acta Mol. Basis Dis. 2015, 1852, 1867–1875. [Google Scholar] [CrossRef] [Green Version]
  54. Duan, H.; Ma, B.; Ma, X.; Wang, H.; Ni, Z.; Wang, B.; Li, X.; Jiang, P.; Umar, M.; Li, M. Anti-diabetic activity of recombinant irisin in STZ-induced insulin-deficient diabetic mice. Int. J. Biol. Macromol. 2016, 84, 457–463. [Google Scholar] [CrossRef] [PubMed]
  55. Xin, C.; Liu, J.; Zhang, J.; Zhu, D.; Wang, H.; Xiong, L.; Lee, Y.; Ye, J.; Lian, K.; Xu, C.; et al. Irisin improves fatty acid oxidation and glucose utilization in type 2 diabetes by regulating the AMPK signaling pathway. Int. J. Obes. 2016, 40, 443–451. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, Y.; Li, R.; Meng, Y.; Li, S.; Donelan, W.; Zhao, Y.; Qi, L.; Zhang, M.; Wang, X.; Cui, T.; et al. Irisin Stimulates Browning of White Adipocytes Through Mitogen-Activated Protein Kinase p38 MAP Kinase and ERK MAP Kinase Signaling. Diabetes 2014, 63, 514–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Liu, S.; Du, F.; Li, X.; Wang, M.; Duan, R.; Zhang, J.; Wu, Y.; Zhang, Q. Effects and underlying mechanisms of irisin on the proliferation and apoptosis of pancreatic β cells. PLoS ONE 2017, 12, e0175498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Bahne, E.; Hansen, M.; Brønden, A.; Sonne, D.P.; Vilsbøll, T.; Knop, F.K. Involvement of glucagon-like peptide-1 in the glucose-lowering effect of metformin. Diabetes Obes. Metab. 2016, 18, 955–961. [Google Scholar] [CrossRef] [PubMed]
  59. Liu, J.; Hu, Y.; Zhang, H.; Xu, Y.; Wang, G. Exenatide treatment increases serum irisin levels in patients with obesity and newly diagnosed type 2 diabetes. J. Diabetes Complicat. 2016, 30, 1555–1559. [Google Scholar] [CrossRef]
  60. Drucker, D.J. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab. 2018, 27, 740–756. [Google Scholar] [CrossRef] [Green Version]
  61. Ramracheya, R.; Chapman, C.; Chibalina, M.; Dou, H.; Miranda, C.; González, A.; Moritoh, Y.; Shigeto, M.; Zhang, Q.; Braun, M.; et al. GLP-1 suppresses glucagon secretion in human pancreatic alpha-cells by inhibition of P/Q-type Ca2+ channels. Physiol. Rep. 2018, 6, e13852. [Google Scholar] [CrossRef]
  62. Lee, Y.-S.; Jun, H.-S. Anti-diabetic actions of glucagon-like peptide-1 on pancreatic beta-cells. Metabolism 2014, 63, 9–19. [Google Scholar] [CrossRef]
  63. Natalicchio, A.; Biondi, G.; Marrano, N.; Labarbuta, R.; Tortosa, F.; Spagnuolo, R.; D’Oria, R.; Carchia, E.; Leonardini, A.; Cignarelli, A.; et al. Long-Term Exposure of Pancreatic β-Cells to Palmitate Results in SREBP-1C-Dependent Decreases in GLP-1 Receptor Signaling via CREB and AKT and Insulin Secretory Response. Endocrinology 2016, 157, 2243–2258. [Google Scholar] [CrossRef] [Green Version]
  64. Natalicchio, A.; de Stefano, F.; Orlando, M.R.; Melchiorre, M.; Leonardini, A.; Cignarelli, A.; Labarbuta, R.; Marchetti, P.; Perrini, S.; Laviola, L.; et al. Exendin-4 Prevents c-Jun N-Terminal Protein Kinase Activation by Tumor Necrosis Factor-α (TNFα) and Inhibits TNFα-Induced Apoptosis in Insulin-Secreting Cells. Endocrinology 2010, 151, 2019–2029. [Google Scholar] [CrossRef]
  65. Natalicchio, A.; Labarbuta, R.; Tortosa, F.; Biondi, G.; Marrano, N.; Peschechera, A.; Carchia, E.; Orlando, M.R.; Leonardini, A.; Cignarelli, A.; et al. Exendin-4 protects pancreatic beta cells from palmitate-induced apoptosis by interfering with GPR40 and the MKK4/7 stress kinase signalling pathway. Diabetologia 2013, 56, 2456–2466. [Google Scholar] [CrossRef] [PubMed]
  66. Natalicchio, A.; Tortosa, F.; Labarbuta, R.; Biondi, G.; Marrano, N.; Carchia, E.; Leonardini, A.; Cignarelli, A.; Bugliani, M.; Marchetti, P.; et al. The p66Shc redox adaptor protein is induced by saturated fatty acids and mediates lipotoxicity-induced apoptosis in pancreatic beta cells. Diabetologia 2015, 58, 1260–1271. [Google Scholar] [CrossRef] [PubMed]
  67. Xu, G.; Kaneto, H.; Lopez-Avalos, M.D.; Weir, G.C.; Bonner-Weir, S. GLP-1/exendin-4 facilitates β-cell neogenesis in rat and human pancreatic ducts. Diabetes Res. Clin. Pr. 2006, 73, 107–110. [Google Scholar] [CrossRef]
  68. Zhang, D.; Xie, T.; Leung, P.S. Irisin Ameliorates Glucolipotoxicity-Associated β-Cell Dysfunction and Apoptosis via AMPK Signaling and Anti-Inflammatory Actions. Cell. Physiol. Biochem. 2018, 51, 924–937. [Google Scholar] [CrossRef] [PubMed]
  69. 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] [PubMed] [Green Version]
  70. Zhang, L.; Li, C.; Zhu, Q.; Li, N.; Zhou, H. Liraglutide, a glucagon-like peptide-1 analog, inhibits high glucose-induced oxidative stress and apoptosis in neonatal rat cardiomyocytes. Exp. Ther. Med. 2019, 17, 3734–3740. [Google Scholar] [CrossRef]
  71. Zhao, S.-M.; Gao, H.-L.; Wang, Y.-L.; Xu, Q.; Guo, C.-Y. Attenuation of High Glucose-Induced Rat Cardiomyocyte Apoptosis by Exendin-4 via Intervention of HO-1/Nrf-2 and the PI3K/AKT Signaling Pathway. Chin. J. Physiol. 2017, 60, 89–96. [Google Scholar] [CrossRef] [Green Version]
  72. Cao, Y.-Y.; Chen, Z.-W.; Gao, Y.-H.; Wang, X.-X.; Ma, J.-Y.; Chang, S.-F.; Qian, J.-Y.; Ge, J.-B. Exenatide Reduces Tumor Necrosis Factor-α-induced Apoptosis in Cardiomyocytes by Alleviating Mitochondrial Dysfunction. Chin. Med. J. 2015, 128, 3211–3218. [Google Scholar] [CrossRef]
  73. 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] [PubMed] [Green Version]
  74. Wu, X.-M.; Ou, Q.-Y.; Zhao, W.; Liu, J.; Zhang, H. The GLP-1 Analogue Liraglutide Protects Cardiomyocytes from High Glucose-induced Apoptosis by Activating the Epac-1/Akt Pathway. Exp. Clin. Endocrinol. Diabetes 2014, 122, 608–614. [Google Scholar] [CrossRef] [PubMed]
  75. Nyström, T.; Gutniak, M.K.; Zhang, Q.; Zhang, F.; Holst, J.J.; Ahrén, B.; Sjöholm, Å. Effects of glucagon-like peptide-1 on endothelial function in type 2 diabetes patients with stable coronary artery disease. Am. J. Physiol. Metab. 2004, 287, E1209–E1215. [Google Scholar] [CrossRef] [PubMed]
  76. Ceriello, A.; Esposito, K.; Testa, R.; Bonfigli, A.R.; Marra, M.; Giugliano, D. The Possible Protective Role of Glucagon-Like Peptide 1 on Endothelium During the Meal and Evidence for an “Endothelial Resistance” to Glucagon-Like Peptide 1 in Diabetes. Diabetes Care 2011, 34, 697–702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Shiraki, A.; Oyama, J.-I.; Komoda, H.; Asaka, M.; Komatsu, A.; Sakuma, M.; Kodama, K.; Sakamoto, Y.; Kotooka, N.; Hirase, T.; et al. The glucagon-like peptide 1 analog liraglutide reduces TNF-α-induced oxidative stress and inflammation in endothelial cells. Atherosclerosis 2012, 221, 375–382. [Google Scholar] [CrossRef]
  78. Nikolaidis, L.A.; Elahi, D.; Hentosz, T.; Doverspike, A.; Huerbin, R.; Zourelias, L.; Stolarski, C.; Shen, Y.-T.; Shannon, R.P. Recombinant Glucagon-Like Peptide-1 Increases Myocardial Glucose Uptake and Improves Left Ventricular Performance in Conscious Dogs with Pacing-Induced Dilated Cardiomyopathy. Circulation 2004, 110, 955–961. [Google Scholar] [CrossRef] [Green Version]
  79. Nikolaidis, L.A.; Doverspike, A.; Hentosz, T.; Zourelias, L.; Shen, Y.-T.; Elahi, D.; Shannon, R.P. Glucagon-Like Peptide-1 Limits Myocardial Stunning following Brief Coronary Occlusion and Reperfusion in Conscious Canines. J. Pharmacol. Exp. Ther. 2004, 312, 303–308. [Google Scholar] [CrossRef] [Green Version]
  80. 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]
  81. Bose, A.K.; Mocanu, M.M.; Carr, R.D.; Brand, C.L.; Yellon, D.M. Glucagon-like Peptide 1 Can Directly Protect the Heart Against Ischemia/Reperfusion Injury. Diabetes 2004, 54, 146–151. [Google Scholar] [CrossRef] [Green Version]
  82. Wohlfart, P.; Linz, W.; Hübschle, T.; Linz, D.; Huber, J.; Hess, S.; Crowther, D.; Werner, U.; Ruetten, H. Cardioprotective effects of lixisenatide in rat myocardial ischemia-reperfusion injury studies. J. Transl. Med. 2013, 11, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Barragan, J.M.; Rodríguez, 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. Metab. 1994, 266, E459–E466. [Google Scholar] [CrossRef]
  84. Barragán, J.M.; E Rodríguez, R.; Eng, J.; Blázquez, E. Interactions of exendin-(9–39) with the effects of glucagon-like peptide-1-(7–36) amide and of exendin-4 on arterial blood pressure and heart rate in rats. Regul. Pept. 1996, 67, 63–68. [Google Scholar] [CrossRef]
  85. 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] [PubMed]
  86. Hirata, K.; Kume, S.; Araki, S.-I.; Sakaguchi, M.; Chin-Kanasaki, M.; Isshiki, K.; Sugimoto, T.; Nishiyama, A.; Koya, D.; Haneda, M.; et al. Exendin-4 has an anti-hypertensive effect in salt-sensitive mice model. Biochem. Biophys. Res. Commun. 2009, 380, 44–49. [Google Scholar] [CrossRef]
  87. Yu, M.; Moreno, C.; Hoagland, K.M.; Dahly, A.; Ditter, K.; Mistry, M.; Roman, R.J. Antihypertensive effect of glucagon-like peptide 1 in Dahl salt-sensitive rats. J. Hypertens. 2003, 21, 1125–1135. [Google Scholar] [CrossRef]
  88. Sun, F.; Wu, S.; Guo, S.; Yu, K.; Yang, Z.; Li, L.; Zhang, Y.; Quan, X.; Ji, L.; Zhan, S. Impact of GLP-1 receptor agonists on blood pressure, heart rate and hypertension among patients with type 2 diabetes: A systematic review and network meta-analysis. Diabetes Res. Clin. Pr. 2015, 110, 26–37. [Google Scholar] [CrossRef]
  89. Kumarathurai, P.; Anholm, C.; Larsen, B.S.; Olsen, R.H.; Madsbad, S.; Kristiansen, O.; Nielsen, O.W.; Haugaard, S.B.; Sajadieh, A. Effects of Liraglutide on Heart Rate and Heart Rate Variability: A Randomized, Double-Blind, Placebo-Controlled Crossover Study. Diabetes Care 2016, 40, 117–124. [Google Scholar] [CrossRef] [Green Version]
  90. Zilov, A.V.; Abdelaziz, S.I.; Alshammary, A.; Al Zahrani, A.; Amir, A.; Khalil, S.H.A.; Brand, K.; Elkafrawy, N.; Hassoun, A.A.; Jahed, A.; et al. Mechanisms of action of metformin with special reference to cardiovascular protection. Diabetes Metab. Res. Rev. 2019, 35, e3173. [Google Scholar] [CrossRef] [Green Version]
  91. Ussher, J.R.; Drucker, D.J. Cardiovascular Actions of Incretin-Based Therapies. Circ. Res. 2014, 114, 1788–1803. [Google Scholar] [CrossRef] [Green Version]
  92. Wang, R.; Lu, L.; Guo, Y.; Lin, F.; Chen, H.; Chen, W.; Chen, M. Effect of Glucagon-like Peptide-1 on High-Glucose-induced Oxidative Stress and Cell Apoptosis in Human Endothelial Cells and Its Underlying Mechanism. J. Cardiovasc. Pharmacol. 2015, 66, 135–140. [Google Scholar] [CrossRef]
  93. Aronis, K.N.; Chamberland, J.P.; Mantzoros, C.S. GLP-1 promotes angiogenesis in human endothelial cells in a dose-dependent manner, through the Akt, Src and PKC pathways. Metabolism 2013, 62, 1279–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Kang, H.-M.; Kang, Y.; Chun, H.J.; Jeong, J.-W.; Park, C. Evaluation of the in vitro and in vivo angiogenic effects of exendin-4. Biochem. Biophys. Res. Commun. 2013, 434, 150–154. [Google Scholar] [CrossRef] [PubMed]
  95. Tashiro, Y.; Sato, K.; Watanabe, T.; Nohtomi, K.; Terasaki, M.; Nagashima, M.; Hirano, T. A glucagon-like peptide-1 analog liraglutide suppresses macrophage foam cell formation and atherosclerosis. Peptides 2014, 54, 19–26. [Google Scholar] [CrossRef]
  96. Gaspari, T.; Welungoda, I.; E Widdop, R.; Simpson, R.W.; E Dear, A. The GLP-1 receptor agonist liraglutide inhibits progression of vascular disease via effects on atherogenesis, plaque stability and endothelial function in an ApoE−/− mouse model. Diabetes Vasc. Dis. Res. 2013, 10, 353–360. [Google Scholar] [CrossRef]
  97. Hirano, T.; Mori, Y. Anti-atherogenic and anti-inflammatory properties of glucagon-like peptide-1, glucose-dependent insulinotropic polypepide, and dipeptidyl peptidase-4 inhibitors in experimental animals. J. Diabetes Investig. 2016, 7, 80–86. [Google Scholar] [CrossRef]
  98. Koshibu, M.; Mori, Y.; Saito, T.; Kushima, H.; Hiromura, M.; Terasaki, M.; Takada, M.; Fukui, T.; Hirano, T. Antiatherogenic effects of liraglutide in hyperglycemic apolipoprotein E-null mice via AMP-activated protein kinase-independent mechanisms. Am. J. Physiol. Metab. 2019, 316, E895–E907. [Google Scholar] [CrossRef]
  99. Rizzo, M.; Nikolic, D.; Patti, A.M.; Mannina, C.; Montalto, G.; McAdams, B.S.; Rizvi, A.A.; Cosentino, F. GLP-1 receptor agonists and reduction of cardiometabolic risk: Potential underlying mechanisms. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 2814–2821. [Google Scholar] [CrossRef]
  100. Leonardini, A.; D’Oria, R.; Incalza, M.A.; Caccioppoli, C.; Buccheri, V.A.; Cignarelli, A.; Paparella, D.; Margari, V.; Natalicchio, A.; Perrini, S.; et al. GLP-1 Receptor Activation Inhibits Palmitate-Induced Apoptosis via Ceramide in Human Cardiac Progenitor Cells. J. Clin. Endocrinol. Metab. 2017, 102, 4136–4147. [Google Scholar] [CrossRef]
  101. Laviola, L.; Leonardini, A.; Melchiorre, M.; Orlando, M.R.; Peschechera, A.; Bortone, A.; Paparella, D.; Natalicchio, A.; Perrini, S.; Giorgino, F. Glucagon-Like Peptide-1 Counteracts Oxidative Stress-Dependent Apoptosis of Human Cardiac Progenitor Cells by Inhibiting the Activation of the c-Jun N-terminal Protein Kinase Signaling Pathway. Endocrinology 2012, 153, 5770–5781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Xin, C.; Zhang, Z.; Gao, G.; Ding, L.; Yang, C.; Wang, C.; Liu, Y.; Guo, Y.; Yang, X.; Zhang, L.; et al. Irisin Attenuates Myocardial Ischemia/Reperfusion Injury and Improves Mitochondrial Function Through AMPK Pathway in Diabetic Mice. Front. Pharmacol. 2020, 11. [Google Scholar] [CrossRef]
  103. Wang, Z.; Chen, K.; Han, Y.; Zhu, H.; Zhou, X.; Tan, T.; Zeng, J.; Zhang, J.; Liu, Y.; Li, Y.; et al. Irisin Protects Heart Against Ischemia-Reperfusion Injury Through a SOD2-Dependent Mitochondria Mechanism. J. Cardiovasc. Pharmacol. 2018, 72, 259–269. [Google Scholar] [CrossRef]
  104. Wang, H.; Zhao, Y.T.; Zhang, S.; Dubielecka, P.M.; Du, J.; Yano, N.; Chin, Y.E.; Zhuang, S.; Qin, G.; Zhao, T.C. Irisin plays a pivotal role to protect the heart against ischemia and reperfusion injury. J. Cell. Physiol. 2017, 232, 3775–3785. [Google Scholar] [CrossRef]
  105. Wang, J.; Zhao, Y.T.; Zhang, L.; Dubielecka, P.M.; Zhuang, S.; Qin, G.; Chin, Y.E.; Zhang, S.; Zhao, T.C. Irisin Improves Myocardial Performance and Attenuates Insulin Resistance in Spontaneous Mutation (Leprdb) Mice. Front. Pharmacol. 2020, 11. [Google Scholar] [CrossRef]
  106. Liao, Q.; Qu, S.; Tang, L.-X.; Li, L.-P.; He, D.-F.; Zeng, C.Y.; Wang, W.E. Irisin exerts a therapeutic effect against myocardial infarction via promoting angiogenesis. Acta Pharmacol. Sin. 2019, 40, 1314–1321. [Google Scholar] [CrossRef]
  107. Moscoso, I.; Márquez, M.C.; Mañero, M.R.; González-Juanatey, J.R.; Lage, R. FNDC5/Irisin counteracts lipotoxic-induced apoptosis in hypoxic H9c2 cells. J. Mol. Endocrinol. 2019, 63, 151–159. [Google Scholar] [CrossRef]
  108. Irisin Attenuates H2O2-Induced Apoptosis in Cardiomyocytes via MicroRNA-19b/AKT/mTOR Signaling Pathway—PubMed n.d. Available online: https://pubmed.ncbi.nlm.nih.gov/31966617/ (accessed on 26 October 2020).
  109. Fan, J.; Zhu, Q.; Wu, Z.; Ding, J.; Qin, S.; Liu, H.; Miao, P. Protective effects of irisin on hypoxia-reoxygenation injury in hyperglycemia-treated cardiomyocytes: Role of AMPK pathway and mitochondrial protection. J. Cell. Physiol. 2020, 235, 1165–1174. [Google Scholar] [CrossRef]
  110. Zhao, Y.T.; Wang, H.; Zhang, S.; Du, J.; Zhuang, S.; Zhao, T.C. Irisin Ameliorates Hypoxia/Reoxygenation-Induced Injury through Modulation of Histone Deacetylase 4. PLoS ONE 2016, 11, e0166182. [Google Scholar] [CrossRef] [Green Version]
  111. Zhang, X.; Hu, C.; Kong, C.-Y.; Song, P.; Wu, H.-M.; Xu, S.-C.; Yuan, Y.-P.; Deng, W.; Ma, Z.-G.; Tang, Q.-Z. FNDC5 alleviates oxidative stress and cardiomyocyte apoptosis in doxorubicin-induced cardiotoxicity via activating AKT. Cell Death Differ. 2020, 27, 540–555. [Google Scholar] [CrossRef] [Green Version]
  112. Li, R.; Wang, X.; Wu, S.; Wu, Y.; Chen, H.; Xin, J.; Li, H.; Lan, J.; Xue, K.; Li, X.; et al. Irisin ameliorates angiotensin II-induced cardiomyocyte apoptosis through autophagy. J. Cell. Physiol. 2019, 234, 17578–17588. [Google Scholar] [CrossRef]
  113. Chen, R.-R.; Fan, X.-H.; Chen, G.; Zeng, G.-W.; Xue, Y.-G.; Liu, X.-T.; Wang, C.-Y. Irisin attenuates angiotensin II-induced cardiac fibrosis via Nrf2 mediated inhibition of ROS/ TGFβ1/Smad2/3 signaling axis. Chem. Interact. 2019, 302, 11–21. [Google Scholar] [CrossRef]
  114. Li, R.-L.; Wu, S.-S.; Wu, Y.; Wang, X.-X.; Chen, H.-Y.; Xin, J.-J.; Li, H.; Lan, J.; Xue, K.-Y.; Li, X.; et al. Irisin alleviates pressure overload-induced cardiac hypertrophy by inducing protective autophagy via mTOR-independent activation of the AMPK-ULK1 pathway. J. Mol. Cell. Cardiol. 2018, 121, 242–255. [Google Scholar] [CrossRef] [PubMed]
  115. Xin, T.; Lu, C. Irisin activates Opa1-induced mitophagy to protect cardiomyocytes against apoptosis following myocardial infarction. Aging 2020, 12, 4474–4488. [Google Scholar] [CrossRef]
  116. Zhang, M.; Xu, Y.; Jiang, L. Irisin attenuates oxidized low-density lipoprotein impaired angiogenesis through AKT/mTOR/S6K1/Nrf2 pathway. J. Cell. Physiol. 2019, 234, 18951–18962. [Google Scholar] [CrossRef] [PubMed]
  117. Zhao, Y.T.; Wang, J.; Yano, N.; Zhang, L.X.; Wang, H.; Zhang, S.; Qin, G.; Dubielecka, P.M.; Zhuang, S.; Liu, P.Y.; et al. Irisin promotes cardiac progenitor cell-induced myocardial repair and functional improvement in infarcted heart. J. Cell. Physiol. 2019, 234, 1671–1681. [Google Scholar] [CrossRef] [PubMed]
  118. Fu, J.; Han, Y.; Wang, J.; Liu, Y.; Zheng, S.; Zhou, L.; Jose, P.A.; Zeng, C. Irisin Lowers Blood Pressure by Improvement of Endothelial Dysfunction via AMPK-Akt-eNOS-NO Pathway in the Spontaneously Hypertensive Rat. J. Am. Hear. Assoc. 2016, 5. [Google Scholar] [CrossRef] [Green Version]
  119. Han, F.; Zhang, S.; Hou, N.; Wang, D.; Sun, X. Irisin improves endothelial function in obese mice through the AMPK-eNOS pathway. Am. J. Physiol. Circ. Physiol. 2015, 309, H1501–H1508. [Google Scholar] [CrossRef] [Green Version]
  120. Hou, N.; Du, G.; Han, F.; Zhang, J.; Jiao, X.; Sun, X. Irisin Regulates Heme Oxygenase-1/Adiponectin Axis in Perivascular Adipose Tissue and Improves Endothelial Dysfunction in Diet-Induced Obese Mice. Cell. Physiol. Biochem. 2017, 42, 603–614. [Google Scholar] [CrossRef]
  121. Fu, J.; Han, Y.; Wang, J.; Jose, P.; Zeng, C. Irisin Lowered Blood Pressure by Augmenting Acetylcholine-Mediated Vasodilation via AMPK-Akt-eNOS-NO Signal Pathway in the Spontaneously Hypertensive Rat. J. Am. Soc. Hypertens. 2016, 10, e4. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, W.; Chang, L.; Zhang, C.; Zhang, R.; Li, Z.; Chai, B.; Li, J.; Chen, E.; Mulholland, M. Central and Peripheral Irisin Differentially Regulate Blood Pressure. Cardiovasc. Drugs Ther. 2015, 29, 121–127. [Google Scholar] [CrossRef] [Green Version]
  123. Huo, C.-J.; Yu, X.-J.; Sun, Y.-J.; Li, H.-B.; Su, Q.; Bai, J.; Li, Y.; Liu, K.-L.; Qi, J.; Zhou, S.-W.; et al. Irisin lowers blood pressure by activating the Nrf2 signaling pathway in the hypothalamic paraventricular nucleus of spontaneously hypertensive rats. Toxicol. Appl. Pharmacol. 2020, 394, 114953. [Google Scholar] [CrossRef]
  124. Jiang, M.; Wan, F.; Wang, F.; Wu, Q. Irisin relaxes mouse mesenteric arteries through endothelium-dependent and endothelium-independent mechanisms. Biochem. Biophys. Res. Commun. 2015, 468, 832–836. [Google Scholar] [CrossRef] [PubMed]
  125. Brailoiu, E.; Deliu, E.; Sporici, R.A.; Brailoiu, G.C. Irisin evokes bradycardia by activating cardiac-projecting neurons of nucleus ambiguus. Physiol. Rep. 2015, 3, e12419. [Google Scholar] [CrossRef]
  126. Wu, F.; Song, H.; Zhang, Y.; Zhang, Y.; Mu, Q.; Jiang, M.; Wang, F.; Zhang, W.; Li, L.; Li, H.; et al. Irisin Induces Angiogenesis in Human Umbilical Vein Endothelial Cells In Vitro and in Zebrafish Embryos In Vivo via Activation of the ERK Signaling Pathway. PLoS ONE 2015, 10, e0134662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Zhang, Y.; Song, H.; Zhang, Y.; Wu, F.; Mu, Q.; Jiang, M.; Wang, F.; Zhang, W.; Li, L.; Shao, L.; et al. Irisin Inhibits Atherosclerosis by Promoting Endothelial Proliferation Through microRNA126-5p. J. Am. Hear. Assoc. 2016, 5. [Google Scholar] [CrossRef] [Green Version]
  128. Song, H.; Shiwu, L.; Zhang, Y.; Zhang, Y.; Wang, F.; Jiang, M.; Wang, Z.; Zhang, M.; Li, W.X.; Yang, L.; et al. Irisin Promotes Human Umbilical Vein Endothelial Cell Proliferation through the ERK Signaling Pathway and Partly Suppresses High Glucose-Induced Apoptosis. PLoS ONE 2014, 9, e110273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Zhu, D.; Wang, H.; Zhang, J.; Zhang, X.; Xin, C.; Zhang, F.; Lee, Y.; Zhang, L.; Lian, K.; Yan, W.; et al. Irisin improves endothelial function in type 2 diabetes through reducing oxidative/nitrative stresses. J. Mol. Cell. Cardiol. 2015, 87, 138–147. [Google Scholar] [CrossRef]
  130. Deng, X.; Huang, W.; Peng, J.; Zhu, T.-T.; Sun, X.-L.; Zhou, X.-Y.; Yang, H.; Xiong, J.-F.; He, H.-Q.; Xu, Y.-H.; et al. Irisin Alleviates Advanced Glycation End Products-Induced Inflammation and Endothelial Dysfunction via Inhibiting ROS-NLRP3 Inflammasome Signaling. Inflammation 2017, 41, 260–275. [Google Scholar] [CrossRef]
  131. Zhu, G.; Wang, J.; Song, M.; Zhou, F.; Fu, D.; Ruan, G.; Zhu, X.; Bai, Y.; Huang, L.; Pang, R.; et al. Irisin Increased the Number and Improved the Function of Endothelial Progenitor Cells in Diabetes Mellitus Mice. J. Cardiovasc. Pharmacol. 2016, 68, 67–73. [Google Scholar] [CrossRef] [Green Version]
  132. Zhang, Y.; Mu, Q.; Zhou, Z.; Song, H.; Zhang, Y.; Wu, F.; Jiang, M.; Wang, F.; Zhang, W.; Li, L.; et al. Protective Effect of Irisin on Atherosclerosis via Suppressing Oxidized Low-Density Lipoprotein Induced Vascular Inflammation and Endothelial Dysfunction. PLoS ONE 2016, 11, e0158038. [Google Scholar] [CrossRef] [PubMed]
  133. Lu, J.; Xiang, G.; Liu, M.; Mei, W.; Xiang, L.; Dong, J. Irisin protects against endothelial injury and ameliorates atherosclerosis in apolipoprotein E-Null diabetic mice. Atherosclerosis 2015, 243, 438–448. [Google Scholar] [CrossRef]
  134. De Silva, A.; Salem, V.; Long, C.J.; Makwana, A.; Newbould, R.D.; Rabiner, E.A.; Ghatei, M.A.; Bloom, S.R.; Matthews, P.M.; Beaver, J.D.; et al. The Gut Hormones PYY3-36 and GLP-17-36 amide Reduce Food Intake and Modulate Brain Activity in Appetite Centers in Humans. Cell Metab. 2011, 14, 700–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Schlögl, H.; Kabisch, S.; Horstmann, A.; Lohmann, G.; Müller, K.; Lepsien, J.; Busse-Voigt, F.; Kratzsch, J.; Pleger, B.; Villringer, A.; et al. Exenatide-Induced Reduction in Energy Intake Is Associated with Increase in Hypothalamic Connectivity. Diabetes Care 2013, 36, 1933–1940. [Google Scholar] [CrossRef] [Green Version]
  136. Kulve, J.S.T.; Veltman, D.J.; van Bloemendaal, L.; Barkhof, F.; Drent, M.L.; Diamant, M.; Ijzerman, R.G. Liraglutide Reduces CNS Activation in Response to Visual Food Cues Only After Short-term Treatment in Patients with Type 2 Diabetes. Diabetes Care 2015, 39, 214–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Van Bloemendaal, L.; Veltman, D.J.; Kulve, J.S.T.; Drent, M.L.; Barkhof, F.; Diamant, M.; Ijzerman, R.G. Emotional eating is associated with increased brain responses to food-cues and reduced sensitivity to GLP-1 receptor activation. Obesity 2015, 23, 2075–2082. [Google Scholar] [CrossRef]
  138. Van Bloemendaal, L.; Ijzerman, R.G.; Kulve, J.S.T.; Barkhof, F.; Konrad, R.J.; Drent, M.L.; Veltman, D.J.; Diamant, M. GLP-1 Receptor Activation Modulates Appetite- and Reward-Related Brain Areas in Humans. Diabetes 2014, 63, 4186–4196. [Google Scholar] [CrossRef] [Green Version]
  139. Secher, A.; Jelsing, J.; Baquero, A.F.; Hecksher-Sørensen, J.; Cowley, M.A.; Dalbøge, L.S.; Hansen, G.; Grove, K.L.; Pyke, C.; Raun, K.; et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J. Clin. Investig. 2014, 124, 4473–4488. [Google Scholar] [CrossRef] [Green Version]
  140. Sisley, S.; Gutierrez-Aguilar, R.; Scott, M.; D’Alessio, D.A.; Sandoval, D.A.; Seeley, R.J. Neuronal GLP1R mediates liraglutide’s anorectic but not glucose-lowering effect. J. Clin. Investig. 2014, 124, 2456–2463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Kanoski, S.E.; Fortin, S.M.; Arnold, M.; Grill, H.J.; Hayes, M.R. Peripheral and Central GLP-1 Receptor Populations Mediate the Anorectic Effects of Peripherally Administered GLP-1 Receptor Agonists, Liraglutide and Exendin-4. Endocrinology 2011, 152, 3103–3112. [Google Scholar] [CrossRef] [Green Version]
  142. Hunter, K.; Hölscher, C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012, 13, 33. [Google Scholar] [CrossRef] [Green Version]
  143. Kastin, A.J.; Akerstrom, V. Entry of exendin-4 into brain is rapid but may be limited at high doses. Int. J. Obes. 2003, 27, 313–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Larsen, P.; Tang-Christensen, M.; Holst, J.J.; Ørskov, C. Distribution of glucagon-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuroscience 1997, 77, 257–270. [Google Scholar] [CrossRef]
  145. Vrang, N.; Larsen, P.J. Preproglucagon derived peptides GLP-1, GLP-2 and oxyntomodulin in the CNS: Role of peripherally secreted and centrally produced peptides. Prog. Neurobiol. 2010, 92, 442–462. [Google Scholar] [CrossRef] [PubMed]
  146. Nian, M.; Drucker, D.J.; Irwin, D. Divergent regulation of human and rat proglucagon gene promoters in vivo. Am. J. Physiol. Content 1999, 277, G829–G837. [Google Scholar] [CrossRef]
  147. Natalicchio, A.; Marrano, N.; Biondi, G.; DiPaola, L.; Spagnuolo, R.; Cignarelli, A.; Perrini, S.; Laviola, L.; Giorgino, F. Irisin increases the expression of anorexigenic and neurotrophic genes in mouse brain. Diabetes Metab. Res. Rev. 2019, 36. [Google Scholar] [CrossRef]
  148. Butt, Z.D.; Hackett, J.D.; Volkoff, H. Irisin in goldfish (Carassius auratus): Effects of irisin injections on feeding behavior and expression of appetite regulators, uncoupling proteins and lipoprotein lipase, and fasting-induced changes in FNDC5 expression. Peptides 2017, 90, 27–36. [Google Scholar] [CrossRef]
  149. Ferrante, C.; Orlando, G.; Recinella, L.; Leone, S.; Chiavaroli, A.; di Nisio, C.; Shohreh, R.; Manippa, F.; Ricciuti, A.; Vacca, M.; et al. Central inhibitory effects on feeding induced by the adipo-myokine irisin. Eur. J. Pharmacol. 2016, 791, 389–394. [Google Scholar] [CrossRef]
  150. Tekin, S.; Erden, Y.; Ozyalin, F.; Cigremis, Y.; Colak, C.; Sandal, S. The effects of intracerebroventricular infusion of irisin on feeding behaviour in rats. Neurosci. Lett. 2017, 645, 25–32. [Google Scholar] [CrossRef]
  151. Kim, O.Y.; Song, J. The Role of Irisin in Alzheimer’s Disease. J. Clin. Med. 2018, 7, 407. [Google Scholar] [CrossRef] [Green Version]
  152. Young, M.F.; Valaris, S.; Wrann, C.D. A role for FNDC5/Irisin in the beneficial effects of exercise on the brain and in neurodegenerative diseases. Prog. Cardiovasc. Dis. 2019, 62, 172–178. [Google Scholar] [CrossRef]
  153. Zhang, J.; Zhang, W. Can irisin be a linker between physical activity and brain function? Biomol. Concepts 2016, 7, 253–258. [Google Scholar] [CrossRef]
  154. Ruan, Q.; Zhang, L.; Ruan, J.; Zhang, X.; Chen, J.; Ma, C.; Yu, Z. Detection and quantitation of irisin in human cerebrospinal fluid by tandem mass spectrometry. Peptides 2018, 103, 60–64. [Google Scholar] [CrossRef] [PubMed]
  155. Perry, T.; Lahiri, D.K.; Chen, D.; Zhou, J.; Shaw, K.T.Y.; Egan, J.M.; Greig, N.H. A Novel Neurotrophic Property of Glucagon-Like Peptide 1: A Promoter of Nerve Growth Factor-Mediated Differentiation in PC12 Cells. J. Pharmacol. Exp. Ther. 2002, 300, 958–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Perry, T.; Haughey, N.J.; Mattson, M.P.; Egan, J.M.; Greig, N.H. Protection and Reversal of Excitotoxic Neuronal Damage by Glucagon-Like Peptide-1 and Exendin-4. J. Pharmacol. Exp. Ther. 2002, 302, 881–888. [Google Scholar] [CrossRef] [Green Version]
  157. During, M.J.; Cao, L.; Zuzga, D.S.; Francis, J.S.; Fitzsimons, H.L.; Jiao, X.; Bland, R.J.; Klugmann, M.; A Banks, W.; Drucker, D.J.; et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat. Med. 2003, 9, 1173–1179. [Google Scholar] [CrossRef]
  158. Nguyen, T.T.; Ta, Q.T.H.; Nguyen, T.K.O.; Nguyen, T.T.D.; van Giau, V.; Vo, V.G.; Nguyen, T.; Nguyen, T. Type 3 Diabetes and Its Role Implications in Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 3165. [Google Scholar] [CrossRef]
  159. Batista, A.F.; Bodart-Santos, V.; de Felice, F.G.; Ferreira, S.T. Neuroprotective Actions of Glucagon-Like Peptide-1 (GLP-1) Analogues in Alzheimer’s and Parkinson’s Diseases. CNS Drugs 2019, 33, 209–223. [Google Scholar] [CrossRef]
  160. Chen, S.; Sun, J.; Zhao, G.; Guo, A.; Chen, Y.; Fu, R.; Deng, Y. Liraglutide Improves Water Maze Learning and Memory Performance While Reduces Hyperphosphorylation of Tau and Neurofilaments in APP/PS1/Tau Triple Transgenic Mice. Neurochem. Res. 2017, 42, 2326–2335. [Google Scholar] [CrossRef]
  161. Hansen, H.H.; Fabricius, K.; Barkholt, P.; Niehoff, M.L.; Morley, J.E.; Jelsing, J.; Pyke, C.; Knudsen, L.B.; Farr, S.A.; Vrang, N. The GLP-1 Receptor Agonist Liraglutide Improves Memory Function and Increases Hippocampal CA1 Neuronal Numbers in a Senescence-Accelerated Mouse Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2015, 46, 877–888. [Google Scholar] [CrossRef] [Green Version]
  162. McClean, P.L.; Parthsarathy, V.; Faivre, E.; Hölscher, C. The Diabetes Drug Liraglutide Prevents Degenerative Processes in a Mouse Model of Alzheimer’s Disease. J. Neurosci. 2011, 31, 6587–6594. [Google Scholar] [CrossRef]
  163. Lourenco, M.V.; Frozza, R.L.; de Freitas, G.B.; Zhang, H.; Kincheski, G.C.; Ribeiro, F.C.; Gonçalves, R.A.; Clarke, J.R.; Beckman, D.; Staniszewski, A.; et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models. Nat. Med. 2019, 25, 165–175. [Google Scholar] [CrossRef]
  164. Peng, H.; Wang, Q.; Lou, T.; Qin, J.; Jung, S.; Shetty, V.; Yanlin, W.; Wang, Y.; Feng, X.-H.; Mitch, W.E.; et al. Myokine mediated muscle-kidney crosstalk suppresses metabolic reprogramming and fibrosis in damaged kidneys. Nat. Commun. 2017, 8, 1–15. [Google Scholar] [CrossRef] [Green Version]
  165. Zhang, R.; Ji, J.; Zhou, X.; Li, R. Irisin Pretreatment Protects Kidneys against Acute Kidney Injury Induced by Ischemia/Reperfusion via Upregulating the Expression of Uncoupling Protein 2. BioMed Res. Int. 2020, 2020, 1–13. [Google Scholar] [CrossRef]
  166. Liu, Y.; Fu, Y.; Liu, Z.; Shu, S.; Wang, Y.; Cai, J.; Ang, C.T.; Dong, Z. Irisin is induced in renal ischemia-reperfusion to protect against tubular cell injury via suppressing p53. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165792. [Google Scholar] [CrossRef]
  167. Jin, Y.; Li, Z.; Jiango, X.; Wu, F.; Li, Z.; Chen, H. Irisin alleviates renal injury caused by sepsis via the NF-κB signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 6470–6476. [Google Scholar] [CrossRef]
  168. Zhang, J.; Bi, J.; Ren, Y.; Du, Z.; Li, T.; Wang, T.; Zhang, L.; Wang, M.; Wei, S.; Lv, Y.; et al. Involvement of GPX4 in irisin’s protection against ischemia reperfusion-induced acute kidney injury. J. Cell. Physiol. 2021, 236, 931–945. [Google Scholar] [CrossRef] [PubMed]
  169. Green, C.J.; Henriksen, T.I.; Pedersen, B.K.; Solomon, T.P.J. Glucagon Like Peptide-1-Induced Glucose Metabolism in Differentiated Human Muscle Satellite Cells Is Attenuated by Hyperglycemia. PLoS ONE 2012, 7, e44284. [Google Scholar] [CrossRef] [PubMed]
  170. Andreozzi, F.; Raciti, G.A.; Nigro, C.; Mannino, G.C.; Procopio, T.; Davalli, A.M.; Beguinot, F.; Sesti, G.; Miele, C.; Folli, F. The GLP-1 receptor agonists exenatide and liraglutide activate Glucose transport by an AMPK-dependent mechanism. J. Transl. Med. 2016, 14, 1–13. [Google Scholar] [CrossRef] [Green Version]
  171. Villanueva-Peñacarrillo, M.L.; Márquez, L.; González, N.; Díaz-Miguel, M.; Valverde, I. Effect of GLP-1 on lipid metabolism in human adipocytes. Horm. Metab. Res. 2001, 33, 73–77. [Google Scholar] [CrossRef]
  172. Challa, T.D.; Beaton, N.; Arnold, M.; Rudofsky, G.; Langhans, W.; Wolfrum, C. Regulation of Adipocyte Formation by GLP-1/GLP-1R Signaling. J. Biol. Chem. 2012, 287, 6421–6430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Young, A.A.; Gedulin, B.R.; Bhavsar, S.; Bodkin, N.; Jodka, C.; Hansen, B.; Denaro, M. Glucose-lowering and insulin-sensitizing actions of exendin-4: Studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 1999, 48, 1026–1034. [Google Scholar] [CrossRef] [PubMed]
  174. Ding, X.; Saxena, N.K.; Lin, S.; Gupta, N.A.; Anania, F.A. Exendin-4, a glucagon-like protein-1 (GLP-1) receptor agonist, reverses hepatic steatosis inob/ob mice. Hepatologia 2005, 43, 173–181. [Google Scholar] [CrossRef] [PubMed]
  175. Fürnsinn, C.; Ebner, K.; Waldhäusl, W. Failure of GLP-l(7–36)amide to affect glycogenesis in rat skeletal muscle. Diabetologia 1995, 38, 864–867. [Google Scholar] [CrossRef]
  176. Nakagawa, Y.; Kawai, K.; Suzuki, H.; Ohashi, S.; Yamashita, K. Glucagon-like peptide-1(7–36) amide and glycogen synthesis in the liver. Diabetologia 1996, 39, 1241–1242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Tang, H.; Yu, R.; Liu, S.; Huwatibieke, B.; Li, Z.; Zhang, W. Irisin Inhibits Hepatic Cholesterol Synthesis via AMPK-SREBP2 Signaling. EBioMedicine 2016, 6, 139–148. [Google Scholar] [CrossRef] [Green Version]
  178. Mo, L.; Shen, J.; Liu, Q.; Zhang, Y.; Kuang, J.; Pu, S.; Cheng, S.; Zou, M.; Jiang, W.; Jiang, C.; et al. Irisin Is Regulated by CAR in Liver and Is a Mediator of Hepatic Glucose and Lipid Metabolism. Mol. Endocrinol. 2016, 30, 533–542. [Google Scholar] [CrossRef] [Green Version]
  179. Park, M.-J.; Kim, D.-I.; Choi, J.-H.; Heo, Y.-R.; Park, S.-H. New role of irisin in hepatocytes: The protective effect of hepatic steatosis in vitro. Cell. Signal. 2015, 27, 1831–1839. [Google Scholar] [CrossRef]
  180. Canivet, C.M.; Bonnafous, S.; Rousseau, D.; LeClere, P.S.; Lacas-Gervais, S.; Patouraux, S.; Sans, A.; Luci, C.; Bailly-Maitre, B.; Iannelli, A.; et al. Hepatic FNDC5 is a potential local protective factor against Non-Alcoholic Fatty Liver. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165705. [Google Scholar] [CrossRef]
  181. Liu, T.-Y.; Xiong, X.-Q.; Ren, X.-S.; Zhao, M.-X.; Shi, C.-X.; Wang, J.-J.; Zhou, Y.-B.; Zhang, F.; Han, Y.; Gao, X.-Y.; et al. FNDC5 Alleviates Hepatosteatosis by Restoring AMPK/mTOR-Mediated Autophagy, Fatty Acid Oxidation, and Lipogenesis in Mice. Diabetes 2016, 65, 3262–3275. [Google Scholar] [CrossRef] [Green Version]
  182. Liu, T.-Y.; Shi, C.-X.; Gao, R.; Sun, H.-J.; Xiong, X.-Q.; Ding, L.; Chen, Q.; Li, Y.-H.; Wang, J.-J.; Kang, Y.-M.; et al. Irisin inhibits hepatic gluconeogenesis and increases glycogen synthesis via the PI3K/Akt pathway in type 2 diabetic mice and hepatocytes. Clin. Sci. 2015, 129, 839–850. [Google Scholar] [CrossRef] [PubMed]
  183. So, W.Y.; Leung, P.S. Irisin ameliorates hepatic glucose/lipid metabolism and enhances cell survival in insulin-resistant human HepG2 cells through adenosine monophosphate-activated protein kinase signaling. Int. J. Biochem. Cell Biol. 2016, 78, 237–247. [Google Scholar] [CrossRef] [PubMed]
  184. Erdogan, M.A.; Yalcin, A. Protective effects of benfotiamine on irisin activity in methotrexate-induced liver injury in rats. Arch. Med. Sci. 2020, 16, 205–211. [Google Scholar] [CrossRef]
  185. Bi, J.; Zhang, J.; Ren, Y.; Du, Z.; Li, Q.; Wang, Y.; Wei, S.; Yang, L.; Zhang, J.; Liu, C.; et al. Irisin alleviates liver ischemia-reperfusion injury by inhibiting excessive mitochondrial fission, promoting mitochondrial biogenesis and decreasing oxidative stress. Redox Biol. 2019, 20, 296–306. [Google Scholar] [CrossRef] [PubMed]
  186. Bi, J.; Yang, L.; Wang, T.; Zhang, J.; Li, T.; Ren, Y.; Wang, M.; Chen, X.; Lv, Y.; Wu, R. Irisin Improves Autophagy of Aged Hepatocytes via Increasing Telomerase Activity in Liver Injury. Oxidative Med. Cell. Longev. 2020, 2020, 1–13. [Google Scholar] [CrossRef]
  187. Reza, M.M.; Subramaniyam, N.; Sim, C.M.; Ge, X.; Sathiakumar, D.; McFarlane, C.; Sharma, M.; Kambadur, R. Irisin is a pro-myogenic factor that induces skeletal muscle hypertrophy and rescues denervation-induced atrophy. Nat. Commun. 2017, 8, 1–17. [Google Scholar] [CrossRef]
  188. Yano, N.; Zhang, L.; Wei, D.; Dubielecka, P.M.; Wei, L.; Zhuang, S.; Zhu, P.; Qin, G.; Liu, P.Y.; Chin, Y.E.; et al. Irisin counteracts high glucose and fatty acid-induced cytotoxicity by preserving the AMPK-insulin receptor signaling axis in C2C12 myoblasts. Am. J. Physiol. Metab. 2020, 318, E791–E805. [Google Scholar] [CrossRef]
  189. Guo, Q.; Wei, X.; Hu, H.; Yang, D.; Zhang, B.; Fan, X.; Liu, J.; He, H.; Oh, Y.; Wu, Q.; et al. The saturated fatty acid palmitate induces insulin resistance through Smad3-mediated down-regulation of FNDC5 in myotubes. Biochem. Biophys. Res. Commun. 2019, 520, 619–626. [Google Scholar] [CrossRef]
  190. Ye, X.; Shen, Y.; Ni, C.; Ye, J.; Xin, Y.; Zhang, W.; Ren, Y. Irisin reverses insulin resistance in C2C12 cells via the p38-MAPK-PGC-1α pathway. Peptides 2019, 119, 170120. [Google Scholar] [CrossRef] [PubMed]
  191. Decreased Irisin Secretion Contributes to Muscle Insulin Resistance in High-Fat Diet Mice—PubMed n.d. Available online: https://pubmed.ncbi.nlm.nih.gov/26261526/ (accessed on 24 June 2020).
  192. Pang, Y.; Zhu, H.; Xu, J.; Yang, L.; Liu, L.; Li, J. β-arrestin-2 is involved in irisin induced glucose metabolism in type 2 diabetes via p38 MAPK signaling. Exp. Cell Res. 2017, 360, 199–204. [Google Scholar] [CrossRef] [PubMed]
  193. Lee, H.J.; Lee, J.O.; Kim, N.; Kim, J.K.; Kim, H.I.; Lee, Y.W.; Kim, S.J.; Choi, J.-I.; Oh, Y.; Kim, J.H.; et al. Irisin, a Novel Myokine, Regulates Glucose Uptake in Skeletal Muscle Cells via AMPK. Mol. Endocrinol. 2015, 29, 873–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Huh, J.Y.; Mougios, V.; Kabasakalis, A.; Fatouros, I.; Siopi, A.; Douroudos, I.I.; Filippaios, A.; Panagiotou, G.; Park, K.H.; Mantzoros, C.S. Exercise-Induced Irisin Secretion Is Independent of Age or Fitness Level and Increased Irisin May Directly Modulate Muscle Metabolism Through AMPK Activation. J. Clin. Endocrinol. Metab. 2014, 99, E2154–E2161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Yang, X.-Y.; Tse, M.C.; Hu, X.; Jia, W.-H.; Du, G.-H.; Chan, C.B. Interaction of CREB and PGC-1α Induces Fibronectin Type III Domain-Containing Protein 5 Expression in C2C12 Myotubes. Cell. Physiol. Biochem. 2018, 50, 1574–1584. [Google Scholar] [CrossRef]
  196. Zhang, Y.; Xie, C.; Wang, H.; Foss, R.M.; Clare, M.; George, E.V.; Li, S.; Katz, A.; Cheng, H.; Ding, Y.; et al. Irisin exerts dual effects on browning and adipogenesis of human white adipocytes. Am. J. Physiol. Metab. 2016, 311, E530–E541. [Google Scholar] [CrossRef] [PubMed]
  197. Huh, J.Y.; Dincer, F.; Mesfum, E.; Mantzoros, C.S. Irisin stimulates muscle growth-related genes and regulates adipocyte differentiation and metabolism in humans. Int. J. Obes. 2014, 38, 1538–1544. [Google Scholar] [CrossRef] [PubMed]
  198. Gao, S.; Li, F.; Li, H.; Huang, Y.; Liu, Y.; Chen, Y. Effects and Molecular Mechanism of GST-Irisin on Lipolysis and Autocrine Function in 3T3-L1 Adipocytes. PLoS ONE 2016, 11, e0147480. [Google Scholar] [CrossRef] [PubMed]
  199. De Oliveira, M.; Mathias, L.S.; Rodrigues, B.M.; Mariani, B.G.; Graceli, J.B.; de Sibio, M.T.; Olimpio, R.M.C.; Moretto, F.C.F.; Deprá, I.C.; Nogueira, C.R. The roles of triiodothyronine and irisin in improving the lipid profile and directing the browning of human adipose subcutaneous cells. Mol. Cell. Endocrinol. 2020, 506, 110744. [Google Scholar] [CrossRef]
  200. Elsen, M.; Raschke, S.; Eckel, J. Browning of white fat: Does irisin play a role in humans? J. Endocrinol. 2014, 222, R25–R38. [Google Scholar] [CrossRef] [Green Version]
  201. Zhao, C.; Liang, J.; Yang, Y.; Yu, M.; Qu, X. The Impact of Glucagon-Like Peptide-1 on Bone Metabolism and Its Possible Mechanisms. Front. Endocrinol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  202. Moayeri, A.; Mohamadpour, M.; Mousavi, S.F.; Shirzadpour, E.; Mohamadpour, S.; Amraei, M. Fracture risk in patients with type 2 diabetes mellitus and possible risk factors: A systematic review and meta-analysis. Ther. Clin. Risk Manag. 2017, 13, 455–468. [Google Scholar] [CrossRef] [Green Version]
  203. Hygum, K.; Starup-Linde, J.; Harsløf, T.; Vestergaard, P.; Langdahl, B.L. MECHANISMS IN ENDOCRINOLOGY: Diabetes mellitus, a state of low bone turnover—A systematic review and meta-analysis. Eur. J. Endocrinol. 2017, 176, R137–R157. [Google Scholar] [CrossRef]
  204. Meng, J.; Ma, X.; Wang, N.; Jia, M.; Bi, L.; Wang, Y.; Li, M.; Zhang, H.; Xue, X.; Hou, Z.; et al. Activation of GLP-1 Receptor Promotes Bone Marrow Stromal Cell Osteogenic Differentiation through β-Catenin. Stem Cell Rep. 2016, 6, 579–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Ma, X.; Meng, J.; Jia, M.; Bi, L.; Zhou, Y.; Wang, Y.; Hu, J.; He, G.; Luo, X. Exendin-4, a Glucagon-Like Peptide-1 Receptor Agonist, Prevents Osteopenia by Promoting Bone Formation and Suppressing Bone Resorption in Aged Ovariectomized Rats. J. Bone Miner. Res. 2013, 28, 1641–1652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Nuche-Berenguer, B.; Moreno, P.; Esbrit, P.; Dapía, S.; Caeiro, J.R.; Cancelas, J.; Haro-Mora, J.J.; Villanueva-Peñacarrillo, M.L. Effect of GLP-1 Treatment on Bone Turnover in Normal, Type 2 Diabetic, and Insulin-Resistant States. Calcif. Tissue Int. 2009, 84, 453–461. [Google Scholar] [CrossRef] [PubMed]
  207. Iepsen, E.W.; Lundgren, J.R.; Hartmann, B.; Pedersen, O.; Hansen, T.; Jørgensen, N.R.; Jensen, J.-E.B.; Holst, J.J.; Madsbad, S.; Torekov, S.S. GLP-1 Receptor Agonist Treatment Increases Bone Formation and Prevents Bone Loss in Weight-Reduced Obese Women. J. Clin. Endocrinol. Metab. 2015, 100, 2909–2917. [Google Scholar] [CrossRef]
  208. Feng, Y.; Su, L.; Zhong, X.; Guohong, W.; Xiao, H.-P.; Li, Y.; Xiu, L.; Wei, G. Exendin-4 promotes proliferation and differentiation of MC3T3-E1 osteoblasts by MAPKs activation. J. Mol. Endocrinol. 2016, 56, 189–199. [Google Scholar] [CrossRef] [Green Version]
  209. Nuche-Berenguer, B.; Moreno, P.; Portal-Núñez, S.; Dapía, S.; Esbrit, P.; Villanueva-Peñacarrillo, M.L. Exendin-4 exerts osteogenic actions in insulin-resistant and type 2 diabetic states. Regul. Pept. 2010, 159, 61–66. [Google Scholar] [CrossRef]
  210. Liu, Z.; Habener, J.F. Glucagon-like Peptide-1 Activation of TCF7L2-dependent Wnt Signaling Enhances Pancreatic Beta Cell Proliferation. J. Biol. Chem. 2008, 283, 8723–8735. [Google Scholar] [CrossRef] [Green Version]
  211. Colaianni, G.; Cinti, S.; Colucci, S.; Grano, M. Irisin and musculoskeletal health. Ann. N. Y. Acad. Sci. 2017, 1402, 5–9. [Google Scholar] [CrossRef]
  212. Colaianni, G.; Cuscito, C.; Mongelli, T.; Oranger, A.; Mori, G.; Brunetti, G.; Colucci, S.; Cinti, S.; Grano, M. Irisin Enhances Osteoblast DifferentiationIn Vitro. Int. J. Endocrinol. 2014, 2014, 1–8. [Google Scholar] [CrossRef] [Green Version]
  213. Colaianni, G.; Cuscito, C.; Mongelli, T.; Pignataro, P.; Buccoliero, C.; Liu, P.; Lu, P.; Sartini, L.; di Comite, M.; Mori, G.; et al. The myokine irisin increases cortical bone mass. Proc. Natl. Acad. Sci. USA 2015, 112, 12157–12162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Qiao, X.Y.; Nie, Y.; Ma, Y.X.; Chen, Y.; Cheng, R.; Yinrg, W.Y.; Hu, Y.; Xu, W.M.; Xu, L.Z. Irisin promotes osteoblast proliferation and differentiation via activating the MAP kinase signaling pathways. Sci. Rep. 2016, 6, 18732. [Google Scholar] [CrossRef] [Green Version]
  215. Polyzos, S.A.; Anastasilakis, A.D.; Efstathiadou, Z.A.; Makras, P.; Perakakis, N.; Kountouras, J.; Mantzoros, C.S. Irisin in metabolic diseases. Endocrine 2018, 59, 260–274. [Google Scholar] [CrossRef] [PubMed]
  216. Adriaenssens, A.E.; Biggs, E.K.; Darwish, T.; Tadross, J.; Sukthankar, T.; Girish, M.; Polex-Wolf, J.; Lam, B.Y.; Zvetkova, I.; Pan, W.; et al. Glucose-Dependent Insulinotropic Polypeptide Receptor-Expressing Cells in the Hypothalamus Regulate Food Intake. Cell Metab. 2019, 30, 987–996.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Samms, R.J.; Coghlan, M.P.; Sloop, K.W. How May GIP Enhance the Therapeutic Efficacy of GLP-1? Trends Endocrinol. Metab. 2020, 31, 410–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Nissen, A.; Christensen, M.; Knop, F.K.; Vilsbøll, T.; Holst, J.J.; Hartmann, B. Glucose-Dependent Insulinotropic Polypeptide Inhibits Bone Resorption in Humans. J. Clin. Endocrinol. Metab. 2014, 99, E2325–E2329. [Google Scholar] [CrossRef]
  219. Skov-Jeppesen, K.; Svane, M.S.; Martinussen, C.; Gabe, M.B.; Gasbjerg, L.S.; Veedfald, S.; Bojsen-Møller, K.N.; Madsbad, S.; Holst, J.J.; Rosenkilde, M.M.; et al. GLP-2 and GIP exert separate effects on bone turnover: A randomized, placebo-controlled, crossover study in healthy young men. Bone 2019, 125, 178–185. [Google Scholar] [CrossRef]
  220. De Fronzo, R.A. Insulin resistance, lipotoxicity, type 2 diabetes and atherosclerosis: The missing links. The Claude Bernard Lecture 2009. Diabetologia 2010, 53, 1270–1287. [Google Scholar] [CrossRef] [Green Version]
  221. Creutzfeldt, M.W. The incretin concept today. Diabetologia 1979, 16, 75–85. [Google Scholar] [CrossRef] [Green Version]
Figure 1. GLP-1 and irisin synthesis and aminoacid sequences. (a) Schematic representation of post-translational proglucagon processing by PC1/3; (b) Schematic representation of FNDC5 structure and cleavage to yield irisin; (c) aminoacid sequences of GLP-1(7–37), GLP-1(7–36)NH2, and irisin. ADAM10, A Disintegrin And Metalloproteinase domain-containing protein 10; C, C-terminal; FNDC5, fibronectin type III domain-containing protein 5; H, hydrophobic domain; GRPP, glicentin-related pancreatic polypeptide; GLP, glucagon-like peptide; IP, intervening peptide; N, N-terminal; PC1/3, prohormone convertase 1/3; SP, signal peptide.
Figure 1. GLP-1 and irisin synthesis and aminoacid sequences. (a) Schematic representation of post-translational proglucagon processing by PC1/3; (b) Schematic representation of FNDC5 structure and cleavage to yield irisin; (c) aminoacid sequences of GLP-1(7–37), GLP-1(7–36)NH2, and irisin. ADAM10, A Disintegrin And Metalloproteinase domain-containing protein 10; C, C-terminal; FNDC5, fibronectin type III domain-containing protein 5; H, hydrophobic domain; GRPP, glicentin-related pancreatic polypeptide; GLP, glucagon-like peptide; IP, intervening peptide; N, N-terminal; PC1/3, prohormone convertase 1/3; SP, signal peptide.
Biomolecules 11 00286 g001
Figure 2. Main GLP-1R and integrin signaling pathways. GLP-1R and integrins, although structurally very different, share numerous intracellular signaling mediators (green boxes), ultimately leading to cell growth and survival. AKT/PKB, protein kinase B; cAMP, cyclic adenosine monophosphate; CREB, cAMP response element-binding protein; Epac2, exchange protein activated by cAMP 2; ERK, extracellular signal-regulated kinases; FAK, focal adhesion kinase; GLP-1, glucagon-like peptide-1; GLP-1R, glucagon-like peptide-1 receptor; Grb2, growth factor receptor-bound protein 2; IRS-2, insulin receptor substrate 2; MEK, mitogen-activated protein kinase; PI-3K, phospatidylinositol-3 kinase; PKA, protein kinase A; PKC, protein kinase C; RAF, rapidly accelerated fibrosarcoma; RAS, rat sarcoma.
Figure 2. Main GLP-1R and integrin signaling pathways. GLP-1R and integrins, although structurally very different, share numerous intracellular signaling mediators (green boxes), ultimately leading to cell growth and survival. AKT/PKB, protein kinase B; cAMP, cyclic adenosine monophosphate; CREB, cAMP response element-binding protein; Epac2, exchange protein activated by cAMP 2; ERK, extracellular signal-regulated kinases; FAK, focal adhesion kinase; GLP-1, glucagon-like peptide-1; GLP-1R, glucagon-like peptide-1 receptor; Grb2, growth factor receptor-bound protein 2; IRS-2, insulin receptor substrate 2; MEK, mitogen-activated protein kinase; PI-3K, phospatidylinositol-3 kinase; PKA, protein kinase A; PKC, protein kinase C; RAF, rapidly accelerated fibrosarcoma; RAS, rat sarcoma.
Biomolecules 11 00286 g002
Figure 3. (a) GLP-1 and irisin effects in pancreatic islets. Blue arrows indicate irisin effects; orange arrows indicate GLP-1 effects; (b) Signaling pathways activated by GLP-1 and irisin in pancreatic β-cells. Green boxes indicate intracellular mediators activated by both GLP-1 and irisin. ACC, Acetyl-CoA carboxylase; AMPKα, adenosine monophosphate-activated protein kinase α; AKT/PKB, protein kinase B; BAD, Bcl-2 associated agonist of cell death; BAX, Bcl-2-like protein 4; Bcl-2, B-cell lymphoma-2; Bcl-xl, B-cell lymphoma-xl; cAMP, cyclic adenosine monophosphate; CREB, cAMP response element-binding protein; Epac2, exchange protein activated by cAMP 2; ERK1/2, extracellular signal-regulated kinases 1/2; FAS, GLK, glucokinase; GLP-1, glucagon-like peptide-1; GLP-1R, glucagon-like peptide-1 receptor; GLUT2, glucose transporter 2; IRS-2, insulin receptor substrate 2; MAPK, mitogen-activated protein kinase; PDX-1, pancreatic and duodenal homeobox 1; PI-3K, phospatidylinositol-3 kinase; PKA, protein kinase A; PKC, protein kinase C; TG, triglycerides.
Figure 3. (a) GLP-1 and irisin effects in pancreatic islets. Blue arrows indicate irisin effects; orange arrows indicate GLP-1 effects; (b) Signaling pathways activated by GLP-1 and irisin in pancreatic β-cells. Green boxes indicate intracellular mediators activated by both GLP-1 and irisin. ACC, Acetyl-CoA carboxylase; AMPKα, adenosine monophosphate-activated protein kinase α; AKT/PKB, protein kinase B; BAD, Bcl-2 associated agonist of cell death; BAX, Bcl-2-like protein 4; Bcl-2, B-cell lymphoma-2; Bcl-xl, B-cell lymphoma-xl; cAMP, cyclic adenosine monophosphate; CREB, cAMP response element-binding protein; Epac2, exchange protein activated by cAMP 2; ERK1/2, extracellular signal-regulated kinases 1/2; FAS, GLK, glucokinase; GLP-1, glucagon-like peptide-1; GLP-1R, glucagon-like peptide-1 receptor; GLUT2, glucose transporter 2; IRS-2, insulin receptor substrate 2; MAPK, mitogen-activated protein kinase; PDX-1, pancreatic and duodenal homeobox 1; PI-3K, phospatidylinositol-3 kinase; PKA, protein kinase A; PKC, protein kinase C; TG, triglycerides.
Biomolecules 11 00286 g003
Figure 4. Pleiotropic effects of GLP-1 and irisin. Blue arrows indicate irisin effects; orange arrows indicate GLP-1 effects; dotted arrows indicate indirect effects.
Figure 4. Pleiotropic effects of GLP-1 and irisin. Blue arrows indicate irisin effects; orange arrows indicate GLP-1 effects; dotted arrows indicate indirect effects.
Biomolecules 11 00286 g004
Figure 5. Glucose-dependent insulinotropic polypeptide (GIP) and irisin similarities and differences. Blue arrows indicate irisin effects; orange arrows indicate GIP effects.
Figure 5. Glucose-dependent insulinotropic polypeptide (GIP) and irisin similarities and differences. Blue arrows indicate irisin effects; orange arrows indicate GIP effects.
Biomolecules 11 00286 g005
Table 1. Effect of dietary nutrients on secretion of GLP-1 and irisin.
Table 1. Effect of dietary nutrients on secretion of GLP-1 and irisin.
NutrientGLP-1 SecretionIrisin Secretion
Carbohydrates+++
Lipids++ 1++ 2
Proteins+N/A
1 Especially unsaturated FAs, 2 Especially saturated FAs, N/A not available.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Marrano, N.; Biondi, G.; Borrelli, A.; Cignarelli, A.; Perrini, S.; Laviola, L.; Giorgino, F.; Natalicchio, A. Irisin and Incretin Hormones: Similarities, Differences, and Implications in Type 2 Diabetes and Obesity. Biomolecules 2021, 11, 286. https://0-doi-org.brum.beds.ac.uk/10.3390/biom11020286

AMA Style

Marrano N, Biondi G, Borrelli A, Cignarelli A, Perrini S, Laviola L, Giorgino F, Natalicchio A. Irisin and Incretin Hormones: Similarities, Differences, and Implications in Type 2 Diabetes and Obesity. Biomolecules. 2021; 11(2):286. https://0-doi-org.brum.beds.ac.uk/10.3390/biom11020286

Chicago/Turabian Style

Marrano, Nicola, Giuseppina Biondi, Anna Borrelli, Angelo Cignarelli, Sebastio Perrini, Luigi Laviola, Francesco Giorgino, and Annalisa Natalicchio. 2021. "Irisin and Incretin Hormones: Similarities, Differences, and Implications in Type 2 Diabetes and Obesity" Biomolecules 11, no. 2: 286. https://0-doi-org.brum.beds.ac.uk/10.3390/biom11020286

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