Next Article in Journal
Contrast-Enhanced Intraoperative Ultrasound Shows Excellent Performance in Improving Intraoperative Decision-Making
Previous Article in Journal
Biomarkers and Data Visualization of Insulin Resistance and Metabolic Syndrome: An Applicable Approach
Previous Article in Special Issue
Personalized Management of Patients with Proliferative Diabetic Vitreoretinopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

How Can Promoting Skeletal Muscle Health and Exercise in Children and Adolescents Prevent Insulin Resistance and Type 2 Diabetes?

by
Valeria Calcaterra
1,2,*,
Vittoria Carlotta Magenes
2,†,
Alice Bianchi
2,†,
Virginia Rossi
2,†,
Alessandro Gatti
3,
Luca Marin
3,
Matteo Vandoni
3 and
Gianvincenzo Zuccotti
2,4
1
Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
2
Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy
3
Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy
4
Department of Biomedical and Clinical Science, University of Milano, 20157 Milano, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Life 2024, 14(9), 1198; https://doi.org/10.3390/life14091198
Submission received: 31 August 2024 / Revised: 16 September 2024 / Accepted: 19 September 2024 / Published: 21 September 2024

Abstract

:
Skeletal muscle secretome, through its paracrine and endocrine functions, contributes to the maintenance and regulation of overall physiological health. We conducted a narrative review on the role of skeletal muscle and exercise in maintaining glucose homeostasis, driving insulin resistance (IR), and preventing type 2 diabetes in pediatric populations, especially in the context of overweight and obesity. Myokines such as interleukin (IL)-6, IL-8, and IL-15, as well as irisin, myonectin, and myostatin, appear to play a crucial role in IR. Skeletal muscle can also become a target of obesity-induced and IR-induced inflammation. In the correlation between muscle, IR, and inflammation, the role of infiltration of the immune cells and the microvasculature may also be considered. It remains unclear which exercise approach is the best; however, combining aerobic exercise with resistance training seems to be the most effective strategy for managing IR, with high-intensity activities offering superior metabolic benefits and long-term adherence. Encouraging daily participation in enjoyable and engaging exercise is key for long-term commitment and effective glucose metabolism management. Promoting physical activity in children and adolescents must be a top priority for public health, not only in terms of individual quality of life and well-being but also for community health.

1. Introduction

Muscles and bones, which form the musculoskeletal system, account for roughly 55% of a healthy adult’s body weight. This system is crucial not only for enabling movement but also for supporting metabolic health. It achieves this by efficiently utilizing, distributing, and delivering nutrients and other essential substrates [1,2,3].
Physical exercise confers benefits across the entire body by reducing fat mass, enhancing insulin sensitivity, and boosting cardiopulmonary capacity, cerebral blood flow, brain oxygenation, and muscle mass and strength [4]. These effects are primarily attributed to metabolic changes triggered by physical activity, which are regulated by molecules responsible for maintaining cellular homeostasis. Specifically, exercise provides metabolic advantages and serves as a potentially effective non-pharmacological strategy against diabetes and cardiovascular diseases [5].
Research indicates that the protective benefits of physical activity against metabolic and cardiovascular disease and other chronic degenerative conditions can be partly attributed to the anti-inflammatory effects of regular exercise. The identification of myokines produced and released by muscles provides a biological basis for understanding how exercise impacts metabolism and reduces inflammation [6,7].
Interestingly, recent studies have shown that muscle exercise can promote the secretion of anti-inflammatory myokines, such as interleukin (IL)-6, IL-15, myostatin, irisin, and myonectin, able to contrast the pro-inflammatory state induced by overweight and obesity and the proper of insulin resistance (IR) [8,9,10].
IR plays a significant role in linking obesity to various metabolic and cardiovascular complications [11]. Pediatric patients with overweight or obesity often exhibit hyperinsulinemia and demonstrate around a 40% reduction in insulin-stimulated glucose uptake compared to children with normal weight. IR is strongly associated with an elevated risk of developing type 2 diabetes (T2D) [12,13,14]. The progression of impaired glucose tolerance in individuals with obesity is tied to the exacerbation of IR and represents a transitional phase in the natural development of T2D [5].
Regular physical exercise enhances muscle glucose uptake and increases the expression of GLUT4, a glucose transporter, in muscle cells, thereby improving insulin sensitivity and reducing the risk of developing insulin resistance and T2D [15,16]. However, it is not clear what type of exercise is most effective in reducing IR in children and adolescents [17].
We propose to present a narrative review on the role of skeletal muscle as an endocrine organ capable of influencing metabolic processes, focusing on the effects of exercise on IR and T2D in pediatrics, particularly in conditions of overweight and obesity. Promoting skeletal muscle health in children and adolescents may be useful to prevent IR and T2D. Implementing healthy lifestyle programs early on provides a more effective strategy for maintaining metabolic health and overall well-being in adulthood.

2. Methods

We conducted a narrative review [18], offering a non-systematic examination of the literature on the role of skeletal muscle and exercise in maintaining glucose homeostasis and driving IR in pediatric populations, especially in the context of overweight and obesity. To narrow the focus of the review, we selected the most pertinent original research articles, clinical trials, meta-analyses, and reviews published in English on this topic up to July 2024. Case reports, case series, and letters were excluded from consideration. Regarding the role of physical exercise on IR and T2D, we considered only the manuscripts that included pediatric patients (<19 years). The search terms included (alone and/or in combination) skeletal muscle, physical activity, insulin resistance, glucose metabolism, exercise, inflammation, adolescents, children, and obesity. For research purposes, we utilized PubMed and Scopus as electronic databases.
The authors screened the abstracts of the available studies (n = 242). These were refined by reviewing abstracts (n = 173) and conducting thorough full-text assessments of pertinent studies (n = 162), which were critically evaluated for inclusion in the manuscript (n = 156). Furthermore, the reference lists of all articles were examined to identify additional relevant studies.

3. Physiology of Muscle Health and the Myokines Network

3.1. Skeletal Muscle as an Endocrine Organ

The idea that the contraction of muscle cells can stimulate the production of humoral factors capable of influencing metabolic processes was first introduced by Goldstein in 1961 [19]. This idea was subsequently validated, and Pedersen et al. pioneered the identification of these molecules as ‘myokines,’ as cytokines that are produced and released by muscle fibers [20]. Since then, the repertoire of recognized myokines has been steadily increasing, and the definition has broadened to encompass any protein secreted by skeletal muscle, whether it functions in an autocrine, paracrine, or endocrine manner through intercellular communication [21,22]. Nevertheless, while it is common practice to use the term “myokine” regardless of the validation of muscle fibers as the origin of the released protein, not all cytokines regulated by exercise are localized within myofibers. Satellite cells, fibroblasts, endothelial cells, and macrophages residing in muscle tissue can also contribute to the release of “myokines” [23].
Several studies on global protein profiling have provided a comprehensive description of the proteins secreted by human myotubes [23,24,25,26,27]. These analyses have revealed that many secreted myokines play a crucial paracrine role in skeletal muscle development and regeneration, extracellular matrix (ECM) organization, and angiogenesis. Additionally, other myokines have been identified to function in an endocrine capacity, primarily involved in regulating glucose and lipid metabolism, as well as neural function [23]. With regard to their impact on the nervous system, the release of these effector molecules during physical activity appears to influence mood, eating behavior, learning, and locomotor activity, also offering neuroprotection in in vivo and in vitro models [4].
On the other hand, physical inactivity fosters an imbalance among these substances, tipping the scale towards a pro-inflammatory state. This, in turn, perpetuates a vicious cycle of sarcopenia, fat accumulation—particularly visceral fat—and the development of various chronic conditions such as cardiovascular disease, T2D, cancer, dementia, and depression, a phenomenon referred to as “the disease of physical inactivity” [28].
Pedersen’s work suggests that during muscle contraction, skeletal muscles release myokines that function hormonally, exerting targeted endocrine on visceral fat [7], providing a biological basis for understanding the role of exercise on metabolism and reducing inflammation [6,7].
McPherron et al. [29] identified myostatin (MSTN) as the first myokine, which is produced and secreted by skeletal muscle. Myostatin is a highly conserved member of the Transforming Growth Factor (TGF)-β superfamily, and it is considered to have the most significant impact on muscle mass and body fat composition among known myokines, and it serves as an inhibitor of muscle mass gain [22,27,29,30]. In fact, inactivation of the myostatin gene (knockout) leads to significant skeletal muscle hypertrophy in mice, cattle, and humans [22]. Schuelke et al. [31] demonstrated that mutations in the human MSTN gene lead to reduced production of mature myostatin, resulting in increased muscle mass and a concomitant reduction in adipose tissue. Moreover, it inhibits bone healing; on the other hand, myostatin inhibitors have been shown to enhance fracture recovery [32]. Furthermore, regular exercise has been shown to reduce myostatin transcript levels in the skeletal muscle of individuals with obesity and impaired glycemic control. A decrease in myostatin mRNA levels has also been observed following acute exercise in healthy individuals. Additionally, MSTN is regulated in vivo by follistatin (FST) and decorin (DCN), which will be discussed later [33].
Several are the cytokines produced and secreted after physical exercise; an increase in cytokine concentration within the muscle interstitial fluid can be observed after 30 min of physical activity [23,34]. The type of physical activity that elicits a greater systemic cytokine response is typically associated with a higher degree of muscle damage, such as downhill running, eccentric exercise, and resistance training, as well as prolonged or high-intensity workouts [23,35].
However, in the case of IL-6, this pattern does not apply: the increase in its expression and release occurs independently of muscle damage [23]. In fact, its production and secretion appear to be regulated by carbohydrate availability and have been proposed as indicators of the muscle’s metabolic state. Low muscle glycogen content prior to exercise results in higher levels of IL-6 and interleukin-8 (IL-8) transcription after exercise. Moreover, carbohydrate ingestion before exercise attenuates the increase in these transcripts [23].
IL-6 plays a key role in both autocrine and paracrine signaling within skeletal muscle, particularly through the activation of the signal transducer and activator of the transcription 3 (STAT3) pathway, which has been observed in human satellite cells following muscle contraction [36]. In mice, IL-6 is crucial for hypertrophic muscle growth and myogenesis [37].
The endocrine effects of exercise-induced IL-6 release are central to the concept of health-promoting myokines. IL-6 enhances insulin-stimulated glucose uptake and oxidation, stimulates lipolysis and fat oxidation, promotes pancreatic β-cell expansion, and improves insulin secretion and glycemic control by stimulating glucagon-like peptide (GLP)-1 release from intestinal L-cells and pancreatic α-cells [4,38,39,40]. In fact, within the skeletal muscle, IL-6 activates AMP-activated protein kinase (AMPK) and/or phosphatidylinositol 3 (PI3K)-kinase pathways to enhance glucose uptake and fat oxidation. Additionally, IL-6 is released into circulation, where it reaches the liver to stimulate glucose production during exercise and the adipose tissue to promote lipolysis [28]. Additionally, IL-6 supports alternative macrophage activation, which is protective against obesity-induced tissue inflammation and insulin resistance [41].
Recent studies also suggest that exercise can reduce tumor size and growth in mice by mobilizing IL-6-dependent natural killer cells [42]. Overall, IL-6 is associated with several beneficial effects of exercise, including improved glycemic control, fat loss, tumor suppression, and muscle mass maintenance.
IL-6 has commonly been considered a pro-inflammatory cytokine, mainly because of its role when released by monocytes and macrophages in reaction to infection. Nevertheless, when IL-6 is secreted by skeletal muscle, it happens without the presence of other inflammatory factors like IL-10 and tumor necrosis factor (TNF)-α. This suggests that the cytokine response induced by physical exercise does not align with typical inflammation pathways [28]. Exercise increases circulating levels of anti-inflammatory cytokines, such as IL-1ra, IL-10, and soluble TNF receptors, which act as natural inhibitors of TNF-α. During exercise, IL-6 itself may exert anti-inflammatory effects, as evidence suggests that this myokine can suppress the synthesis of IL-1 and TNF-α and stimulate the production of IL-1ra and IL-10 [28].
Not only are IL-6 and myostatin regulated by physical activity and involved in post-exercise metabolic regulation, but several other myokines also play critical roles in reducing subcutaneous and visceral fat and enhancing substrate oxidation capacity, both of which are essential for insulin sensitivity [4].
IL-15, a member of the IL-2 superfamily, is another chemokine produced by skeletal muscle [4,28]. IL-15 signaling appears to be involved in the regulation of muscle fiber composition and contractility, acting as an anabolic factor that stimulates muscle growth. Additionally, IL-15 plays a role in lipid metabolism regulation [4,28]. It has been shown to reduce lipid accumulation in preadipocytes and decrease the mass of white adipose tissue. As a result, a negative relationship has been observed between plasma IL-15 levels and total body fat mass, trunk fat mass, and the percentage of body fat in humans, and overexpression of IL-15 in skeletal muscle has been shown to reduce visceral fat in murine models [4,28].
Additional exercise-regulated myokines promote the proliferation of primary human skeletal muscle cells, such as decorin, leukemia inhibitory factor (LIF), and chitinase-3-like protein 1 (CHI3L1) [43,44,45].
Decorin, a small leucine-rich proteoglycan, is synthesized and secreted by human myotubes in response to muscle contraction, with its levels increasing following physical exercise. It has been demonstrated to inhibit myostatin activity in both fibroblasts and myoblasts and also influences the expression of follistatin, another inhibitor of myostatin [33,43]. In fact, mouse studies have shown that overexpression of decorin in skeletal muscle in vivo enhances the expression of the pro-myogenic factor Mighty, which is downregulated by myostatin. Additionally, Myogenic Differentiation 1 (Myod1) and follistatin expression are elevated in response to decorin overexpression [33,46]. In summary, decorin, which is secreted by myotubes in response to exercise, plays a role in regulating muscle hypertrophy and may contribute to the remodeling of skeletal muscle during exercise.
The myokine LIF is part of the IL-6 cytokine superfamily, which comprises structurally and functionally similar proteins known as neuropoietins (or gp130 cytokines) [47]. It is a contraction-induced myokine, acting in an autocrine and/or paracrine fashion [22]. LIF plays a diverse role in biological processes, including promoting platelet production, the proliferation of hematopoietic cells, bone formation, neural survival and development, and the acute-phase response in liver cells [48]. Moreover, it supports satellite cell proliferation [22]. The expression of LIF mRNA is triggered in human skeletal muscle after resistance exercise, and the LIF protein is released by electrically stimulated human myotubes in culture [44]. LIF enhances the proliferation of human myoblasts and triggers the expression of jun-B and c-Myc in human myotubes. In contrast, knocking down the LIF receptor using siRNA led to decreased proliferation [22].
CHI3L1 (chitinase-3-like protein 1) is a glycoprotein composed of 383 amino acids with a molecular mass of 40 kDa [45,49,50]. While it is well-known as an acute-phase protein that increases during inflammatory diseases, particularly sepsis, CHI3L1 also plays several other roles [45,50]. Notably, it is produced and secreted by skeletal muscle in response to acute physical exercise. The CHI3L1/PAR-2 signaling pathway, in turn, stimulates myocyte proliferation, which is crucial for skeletal muscle remodeling in response to training [45,50]. Additionally, CHI3L1 has been linked to enhanced glucose uptake in skeletal muscles through an AMP-activated protein kinase (AMPK)-dependent mechanism and an increase in intracellular calcium levels via PAR2 [49]. Furthermore, CHI3L1 has been found to influence glucose uptake through the PI3K/AKT pathway [49].
Angiopoietin-like protein 4 (ANGPTL4), part of the angiogenin-like protein family, is expressed across a diverse array of tissues, including the liver, adipose tissue, skeletal muscle, placenta, small intestine, brain, thyroid, kidney, spleen, pituitary gland, hypothalamus, and heart. It is upregulated in response to various physiological conditions, including fasting and physical exercise. Following acute resistance and strength training, ANGPTL4 mRNA levels rise in both serum and skeletal muscle. In non-exercising muscle, the increase in ANGPTL4 is mediated by elevated plasma free fatty acids (FFAs) through PPAR activation, likely to prevent fat overload and ensure the supply of fatty acids to active skeletal muscles [51,52,53]. Given that ANGPTL4 regulates lipid metabolism by inhibiting lipoprotein lipase activity and promoting lipolysis in white adipose tissue, it may contribute to insulin resistance by regulating its expression through PPARs (PPARα, PPARδ/β, PPARγ) [54,55]. A study by van der Kolk et al. [56] examined the impact of a mixed meal rich in saturated fatty acids (SFA) on plasma ANGPTL4 levels and its relationship with muscle LPL activity in vivo, aiming to understand the effects of dietary fat quality and the release of ANGPTL4 from skeletal muscle. The study found that ANGPTL4 is secreted from human forearm muscle after a high-SFA meal in postprandial conditions. However, plasma ANGPTL4 levels were not linked to skeletal muscle LPL activity in vivo following the SFA-rich meal. While dietary fat quality does affect plasma ANGPTL4, its impact on short-term lipid metabolism in skeletal muscle remains to be determined [56]. Additionally, ANGPTL4 has recently been implicated in mice as a mediator of pancreatic β-cell hyperplasia [4,53].
Apelin is a peptide produced by skeletal muscles, with its production triggered by muscle contractions [57]. It is a ligand for the G protein-coupled receptor APJ, and it has been shown in rodents to enhance glucose uptake and mitochondrial oxidative capacity in skeletal muscle [58,59,60]. Furthermore, its levels decline with age in both humans and animals, and mice lacking apelin or its receptor (APLNR) experience significant muscle function deterioration as they age [57]. Restoring apelin activity in these aging mice improves muscle function by enhancing energy production, reducing inflammation, and supporting muscle stem cell (MuSC) regeneration. These findings make apelin a potential biomarker for early muscle deterioration and a target for therapies to combat age-related muscle weakness [57]. In related research, Le Moal et al. [61] identified a specific form of apelin (AP-13) that stimulates skeletal muscle endothelial cells (ECs), improving muscle regeneration and strength in mouse models of muscular dystrophy by creating a supportive environment for MuSCs. This research suggests that targeting apelin signaling, particularly through AP-13, may offer a promising strategy for treating muscle stem cell dysfunction and enhancing muscle repair.
Irisin is a recently identified polypeptide hormone, discovered also in human plasma via mass spectrometry [62], and induced by physical exercise and secreted by muscle. It is produced through the proteolytic processing of FNDC5 [63], whose expression in muscle is, in turn, upregulated by PGC-1α. Irisin was discovered as a myokine that promotes the transformation of white adipose tissue into a brown fat-like phenotype [22]. In mice, the injection of adenoviral particles expressing FNDC5 led to a three- to four-fold increase in irisin levels, resulting in the induction of a brown fat-like cell development program in white adipose tissue and a concomitant increase in energy expenditure [22,63]. Baseline plasma levels of irisin were found to increase in response to 10 weeks of regular physical exercise in humans, suggesting a role for irisin in exercise adaptation. Additionally, irisin has been shown to exert antifibrotic effects on the heart, liver, pancreas, and muscle. Specifically, its antifibrotic effect on muscle was demonstrated in a recent study by Wu et al. [64]. In their study examining the impact of irisin on D-galactose (D-gal)-induced skeletal muscle fibroblasts, it was found that irisin or FNDC5 overexpression effectively inhibited or reduced the effects of D-galactose, such as cellular senescence, fibrosis, and redox imbalance. This protective effect was achieved through modulation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway [64].
Myokines produced and secreted in response to physical activity have also been linked to direct cardiovascular benefits, including increased cardiac output and overall blood volume. Notable among these are vascular endothelial growth factor (VEGF), as well as IL-8, which promotes endothelial cell proliferation and capillary tube formation, and CYR61 (CCN1) and CTGF (CCN2), which are associated with the extracellular matrix (ECM) protein group [4].
During physical exercise, skeletal muscle fibers synthesize and secrete VEGF within vesicles. VEGF is a primary angiogenic factor that plays a crucial role in the development of blood vessels, including the capillaries within skeletal muscle tissue. During muscle contraction, VEGF concentration increases within the muscle interstitium, thereby stimulating angiogenic processes [65]. Moreover, VEGF not only enhances blood vessel growth but also plays a significant role in tissue regeneration [66]. On the other hand, CYR61 (CCN1) and CTGF (CCN2) increase in skeletal muscle following exercise, especially following mechanical loading [67]. They are involved in skeletal muscle remodeling after exercise, as they regulate the expression of genes involved in angiogenesis and ECM remodeling.
The myokine brain-derived neurotrophic factor (BDNF), which increases following physical activity, plays a critical role in neural processes by regulating the growth, survival, and maintenance of neurons [28]. This, in turn, influences information processing, learning, and memory. Studies on individuals with Alzheimer’s disease have shown low plasma concentrations of BDNF, and post-mortem analysis of hippocampal samples has revealed reduced BDNF expression [28,68]. Additionally, low blood levels of BDNF have been associated with major depressive disorder, acute coronary syndromes, and type 2 diabetes mellitus [28]. BDNF also influences energy metabolism and adipose tissue by enhancing fat oxidation through an AMPK-dependent pathway in skeletal muscles, leading to a reduction in adipose tissue mass [69].
In Table 1, different myokines and their roles are resumed.

3.2. Skeletal Muscle and Glucose Metabolism

Skeletal muscle uses three main types of substrates for its energy metabolism: glycogen, glucose, and free fatty acids [70]. The specific energy sources used by muscles depend on several factors, including physical conditioning, diet, type, intensity, and duration of exercise [71]. At rest, in fact, the main energy sources are fatty acids, whereas, during exercise, the energy metabolism of skeletal muscle depends on both glucose and free fatty acids.
According to the intensity of exercise, the major energy substrate is glucose during high-intensity isometric exercise, glucose and glycogen in the case of high-intensity submaximal exercise, and both glucose and free fatty acids in the case of low-intensity submaximal exercise. According to the duration of exercise, instead, in the first hour of mild activity, the primary sources of energy are glucose, glycogen, and free fatty acids, whereas in the following hours, the uptake of free fatty acids increases gradually. Therefore, lipid metabolism plays a predominant role at rest, whereas glucose metabolism is crucial for energy generation during exercise [72].
Skeletal muscle accounts for more than 80% of glucose absorption in the post-meal state [73], playing a vital role in regulating blood glucose levels. It absorbs glucose from the extracellular fluid into the cell via specific sugar transport proteins located on the cell membrane [8,74], known as GLUTs (facilitative glucose transporters) and SGLTs (sodium-dependent glucose co-transporters).
There are three main isoforms of GLUTs in skeletal muscle: the most important one is GLUT4, followed by GLUT1 and GLUT3, which are expressed only in fetal and neonatal muscle [8,75]. Other sugar transporter proteins expressed in skeletal muscle include GLUT5, GLUT6, GLUT8, GLUT10, GLUT11, GLUT12, SGLT1, SGLT2, SGLT3, and SGLT4 [76]. In contrast to GLUT1, which is found on the surface of muscle cells, GLUT4 is an intracellular protein that requires stimulation, such as from insulin or physical exercise, to move to the cell surface and assist in glucose absorption [77]. In patients with type 2 diabetes or insulin resistance, the ability of insulin to stimulate skeletal muscle glucose transport is altered [78,79] due to an impaired GLUT4 translocation to the muscle cell surface [80]. Exercise, on the contrary, can stimulate GLUT4 translocation and glucose transport, both in healthy and type 2 diabetes individuals [81].
Once transported into skeletal muscle cells, glucose is phosphorylated by hexokinase to glucose-6-phosphate, and then it can go through four different intracellular pathways, as described in Figure 1.
Glycogen, the stored form of glucose in skeletal muscle, acts as a glucose buffer. At rest, muscle glycogen levels are regulated by the balance between glycogen synthesis and degradation. However, exercise leads to an increase in glycogen synthesis [82]. During this process, glucose-6-phosphate is first converted to glucose-1-phosphate by the enzyme phosphoglucomutase and then to uridine diphosphate glucose (UDP-glucose) by glycogen synthase (GS), forming the multi-branched glucose polymers characteristic of glycogen particles. When cellular energy demands rise, glycogen is broken down into glucose-1-phosphate by glycogen phosphorylase (GP) and subsequently metabolized through glycolysis [76].
Glucose flux through glycolysis plays a fundamental role in generating adenosine triphosphate (ATP), which is the source of energy for skeletal muscle contractile function. Glucose-6-phosphate is converted to fructose-6-phosphate by the enzyme phosphoglucose isomerase and then to fructose-1,6-bisphosphate by the enzyme phosphofructokinase (PFK). After seven other sequential reactions, fructose-1,6-bisphosphate is finally converted to pyruvate. Pyruvate can be reduced to lactate or, on the other hand, can be oxidated to acetyl-CoA by the enzyme pyruvate dehydrogenase (PDH) and further metabolized via the tricarboxylic acid cycle (TCA) and mitochondrial electron transport chain [76]. The complete oxidation of glucose generates 36 molecules of ATP.
The hexosamine pathway is a glucose-utilizing pathway that starts when fructose-6-phosphate is converted to glucosamine-6-phosphate by the enzyme glutamine fructose-6-phosphate transaminase 1 (GFPT1). This biosynthesis pathway determines the production of uridine diphosphate-N-acetyl glucosamine (UDP-GlcNAc), which is a key metabolite for N- or O-linked glycosylation, which modulates protein expression and activity [83].
The last glucose-utilizing pathway in skeletal muscle is the pentose phosphate pathway. Glucose-6-phosphate is converted to 6-phosphogluconolactone by the enzyme glucose-6-phosphate dehydrogenase (G6PD). Then, it is metabolized through a series of reactions in order to generate important metabolites, such as nicotinamide adenine dinucleotide phosphate (NADPH), which is crucial for reductive biosynthesis reactions such as lipogenesis; ribose 5-phosphate, used for nucleotide synthesis; erythrose-4-phosphate, used for aromatic amino acid synthesis; and ribulose 5-phosphate, often measured to check pentose phosphate pathway activity [84]. At rest, the activity of the pentose phosphate pathway is low in skeletal muscle [85] since it is a differentiated cell-type tissue and does not have high biosynthetic demands. On the contrary, studies showed that the activity of the pentose phosphate pathway increases after damage to provide substrates for muscle repair processes [76].

3.3. Key Myokines in the Regulation of Glucose Metabolism

Skeletal muscle, being a key player in the regulation of glucose metabolism, is also considered the primary driver of whole-body IR. If the main metabolic defect occurs at the level of skeletal muscle, restoring glucose homeostasis by addressing IR in the muscle is fundamental and may be sufficient [73].
IR occurs due to the desensitization of muscle to the action of insulin (to elicit glucose uptake), and it leads to unproperly elevated blood glucose levels [86]. Interestingly, IR in skeletal muscle can appear decades before the onset of pancreatic β-cell failure and symptomatic T2D [8,73,87].
IR negatively affects the glucose uptake at the skeletal muscle levels both in terms of timing and in terms of quantity [8,73]. Indeed, in physiological conditions, postprandial glucose uptake—upon insulin release—increases into muscle linearly with time [8,73,88]. Instead, in a condition of IR, insulin action and glucose uptake are delayed, and this leads to a diminished overall glucose uptake by the skeletal muscle [8,73,88]. This has been shown by hyperinsulinemic-euglycemic clamping studies in patients affected by T2D and controls [8,89].
Among the myokines produced by the muscle IL-6, IL-15, and interleukin-8 (IL-8), [1,15,17], as well as irisin, myonectin, and myostatin [1,15,17] appear to play a crucial role in IR. No other exercise-induced molecules were more extensively evaluated, in this context, in the pediatric field.
Specifically, IL-6 is one of the best-known pro-inflammatory cytokines released by skeletal muscle, and it was shown to contribute to the onset of IR [90,91]. Recently, this molecule was shown to also have an anti-inflammatory effect when released in response to exercise [92]. The different effects are due to the activation of different signaling pathways. Specifically, IL-6 acts as a pro-inflammatory molecule through the Nuclear Factor kappa (NFκB) pathway, but it has an anti-inflammatory potential when released through a calcium-dependent Mitogen-activated Protein Kinase (MAPK) signal [92,93]. Hojman et al. investigated the mechanism involved in the exercise-mediated surge in IL-6 and the related lactate production, evaluating muscle activity upon interval-based cycling in healthy young men, swimming exercise in mice, and electrical stimulation in human muscle cells [94]. The authors discovered that IL-6 levels rise with both the intensity and duration of exercise. Additionally, the release of IL-6 during exercise is influenced by protease activity and lactate production [94].
Similarly, IL-15 is a myokine known to be regulated by exercise and muscle contraction [8,95]. It has been associated with obesity and metabolic syndrome [8,95,96]. This molecule is thought to mediate the benefits of exercise, but the precise effect of muscle activity on IL-15 stimulation varies from study to study [97,98,99,100]. Recently, Khalafi et al. performed a systematic review and meta-analysis to investigate whether exercise (acute and chronic) stimulates an increase in circulating IL-15 concentrations in humans [101]. The authors evaluated a total of 27 studies involving 1310 participants and showed that acute exercise increases circulating IL-15 concentrations immediately and one hour after [101]. Instead, chronic exercise does not have a significant effect on IL-15 concentrations [101]. Thus, the results confirm that acute exercise, increasing IL-15 concentrations, has a potential role in improving metabolism in adults [101]. The ongoing debate about IL-15’s effects highlights the need for further research to clarify the precise role of this myokine [8,95].
Unfortunately, studies evaluating exercise-induced interleukins and their role in me-tabolism in children and adolescents are scarce. Thus, even if the results on adults are promising, further works are needed to better understand the role of these molecules in glucidic metabolism in these patients and how these molecules specifically act.
Myostatin was the first myokine identified. It is part of the TGF-β family of proteins [8,9,102], and it is expressed both in skeletal muscle and in adipose tissue [103]. Myostatin negatively regulates muscular mass. Indeed, its ablation was shown to be correlated to muscle hypertrophy [104,105]. Moreover, growth differentiation factor 8 negatively regulates muscular mass [105,106]. Indeed, its ablation was shown to be correlated to muscle hypertrophy [104,105]. Upon exercise, myostatin decreases, and its expression has been correlated to a decrease in plasma levels of glucose, insulin, and IL-6 and a reduction in IR (calculated with the Homeostatic Model Assessment for Insulin Resistance Index, HOMA-IR) [106,107,108]. Further studies are anyway needed to better understand its correlation with glucose metabolism and eventually consider this molecule as a candidate therapeutic target to combat obesity by promoting muscle growth [8].
Irisin expression, upon muscular exercise, is thought to be involved in the beginning of white adipose tissue, causing white adipose tissue to partially transform into the brown adipose tissue phenotype [62,63,109,110]. As brown tissue is associated with increased thermogenesis, this phenomenon can lead to improvements in terms of obesity and glucose homeostasis [63]. Interestingly, Jedrychowski et al. identified and quantitated human irisin in plasma (using mass spectrometry) in individuals undergoing aerobic interval training [62]. The authors showed that circulating irisin levels significantly increased with exercise and that mass spectrometry can be a possible tool to measure it [62].
Reinehr et al. evaluated 40 children with obesity and 20 normal-weight children (of similar age and gender) after a 1-year outpatient intervention program based on exercise, behavior, and nutrition therapy in order to study the relationships between irisin, puberty and IR after weight loss [110]. The authors evidenced that irisin levels were the highest in children with obesity and with impaired glucose tolerance and the lowest in normal weight children. In addition, their longitudinal analyses showed that changes of irisin were associated with entry into puberty and amelioration of glucidic metabolism (change of fasting glucose, and 2-hour glucose in an oral glucose tolerance test), but not with change of BMI [110].
However, there is controversy regarding the increase in serum irisin following muscular activity, both due to the difficulty in detecting irisin in the blood and the scarcity of studies on this subject, particularly in a pediatric context.
Myonectin is a nutrient-responsive myokine released by muscle contraction that mimics insulin’s ability to promote fatty acid cellular uptake response to exercise [109,110,111]. In addition, myonectin can also increase the translocation of the GLUT4 glucose transporter and promote glucose uptake [112]. Lastly, this molecule has been associated with mitochondrial deoxyribonucleic acid (mtDNA) density; specifically, IR, leading to decreased mtDNA levels, can upregulate myonectin [112]. Even in this case, further studies are needed to clearly understand the role of this molecule in this context [8].
Finally, other myokines shown to have a role in glucose metabolism and muscle include BDNF and Decorin [8,10,112]. Moreover, new myokines that respond to exercise (called “exerkines”) have been identified [113]; their effect on IR must be clarified, especially in the pediatric field, before they can be lev-eraged as potential therapeutic targets.
In Figure 2, the properties of the key myokines involved in muscle contraction and in-fluencing glucose metabolism are schematized.
IR is strictly correlated to weight gain, overnutrition, and obesity; it has been shown that when lean and non-diabetic individuals were given a regimen of overnutrition, they developed IR [114]. The link between IR and obesity may be partially explained by the condition of chronic inflammation proper of both conditions [8,115]. Indeed, adipose tissue releases specific pro-inflammatory bioactive molecules (called adipokines) that have been shown to be dysregulated in conditions of obesity, IR, and diabetes [116,117,118].
Myokines are molecules that facilitate communication between skeletal muscle and other organs such as bone, brain, and adipose tissue [8,9]. Moreover, skeletal muscle itself can become a target for inflammation induced by obesity and IR [8]. Both obesity and IR have been found to stimulate the release of specific cytokine hormones from tissues, known as adipomyokines [103]. Unlike adipokines, which are influenced by factors like body fat, muscle myokines are primarily regulated by exercise and muscle contractions [8,9]. The levels of these circulating factors can vary in response to conditions such as obesity, inflammation, and IR [8,9].
Additionally, in conditions of obesity, IR and/or T2D skeletal muscle becomes infiltrated by immune cells (mainly M1-like macrophages and T cells) [119,120]. Moreover, its adipose tissue depots can expand for up to 10% of the total muscle mass [119]. These depots—intramuscular or subcutaneous—are thought to be the main contributors of pro-inflammatory immune cells in the skeletal muscle [119], and they have been correlated to glucose intolerance and IR [1,19,20,21].
Examining the correlation between muscle, IR, and inflammation, it is worth considering also the role of microvasculature [121,122]. Indeed, evidence confirms that muscle microvasculature is a fundamental site of action of insulin, and it regulates insulin delivery and action on myocytes, thus affecting insulin-mediated glucose disposal [121,122]. IR at the microvascular level due to inflammation is an early event in the development of metabolic IR and eventually T2D; thus, research should nowadays focus on this theme in order to find new potential therapeutical targets [121].
In this context, the role of thiazolidinediones (TZDs) has to also be considered. TZDs, such as pioglitazone and rosiglitazone, help to improve insulin sensitivity by targeting peroxisome proliferator-activated receptor gamma (PPARγ) receptors, which are expressed in adipose tissue but also influence muscle metabolism indirectly [123,124]. Indeed, in skeletal muscle, TZDs enhance insulin-mediated glucose uptake by improving the action of insulin signaling pathways, thus reducing IR [123,124].
In women and adolescents with polycystic ovary syndrome (PCOS), IR in skeletal muscle exacerbates hyperinsulinemia, which, in turn, stimulates excess androgen production by the ovaries, worsening reproductive symptoms [125,126]. The use of TZDs in these patients was shown to improve metabolic outcomes by enhancing insulin sensitivity in skeletal muscle [127,128,129]. In young patients affected by PCOS, metformin remains the most effective and cost-efficient therapy to regulate insulin sensitivity in the condition of overweight and/or obesity [129,130]. While there is evidence supporting TZD efficacy in young women and adolescents with PCOS, further studies are necessary to confirm the results obtained and evaluate long-term effects.
Finally, it is noteworthy that the relationship between skeletal muscle and IR appears to be partially influenced by gender. Recently, Liu et al. explored the gender-specific associations between predicted skeletal muscle mass index (pSMI) and the incidence of type 2 diabetes in a longitudinal cohort study of Chinese adults. The study, which utilized the WATCH (West China Adult Health Cohort) database, included Chinese men and women without diabetes at the study’s start. The researchers calculated pSMI, measured blood glucose levels, and collected self-reported medical histories to identify new cases of diabetes [131]. Then, they concluded that in females, a larger muscular mass is associated with a lower risk of T2D; instead, for males, this association is significant only among those with diminished muscle mass [131]. Moreover, Ciarambino et al. conducted a systematic literature search to identify studies evaluating the association between gender and IR [132]. The authors found that IR is generally more prevalent in men compared to premenopausal women. Instead, after menopause, the incidence of IR in women increases and becomes more comparable to that of men [132]. This difference seems to depend both on body fat distribution and on sexual hormones. Indeed, men tend to accumulate more visceral fat, which is strongly associated with IR and metabolic disturbances [132]. Instead, estrogen, the primary female sex hormone, appears to have protective effects on insulin sensitivity. Indeed, women tend to have better insulin sensitivity during their reproductive years due to the presence of estrogen. However, after menopause, estrogen levels decline as insulin sensitivity [132]. Unfortunately, the literature concerning sex differences in the association between skeletal, muscular mass, and glucidic metabolism is scarce in the pediatric field, and further research should be conducted to drive conclusions on this issue.

4. Effects of Exercise Training on Insulin Resistance and Type 2 Diabetes in Pediatrics

Exercise training is considered an important strategy to cope with IR and related metabolic unbalances [17], and this is of fundamental importance as impairment in insulin signaling is correlated to the future development of severe conditions such as T2D and metabolic syndrome, both in adults and in children or adolescents, primarily in the condition of overweight and obesity [133,134]. Exercise was shown to have favorable effects on metabolic indices (such as blood glucose and insulin levels) related to overweight and obesity acting at various levels. Overweight and excessive fat tissue is associated with IR and exercise; decreasing body weight and adipose tissue leads to a decrease in IR [135].
Specifically, with aerobic exercise, alterations in both metabolic and non-metabolic processes contribute to improvements in IR and associated indices [17,136]. Specifically, muscle contracting leads to the translocation of the glucose transporter 4 (GLUT4) to the plasma membrane and increases the density of this transporter in order to increase glucose uptake from the blood into the muscle [137,138,139,140]. The amount of glucose uptake by the skeletal muscle is determined by the activity duration and intensity: increased exercise intensity and time increase glucose uptake [8,137,138]. In addition, muscular activity has been associated with an increase in insulin receptor substrate 1 (IRS1) phosphorylation, leading to improvement of insulin signal transduction; an improvement of beta-cell and pancreatic islets function, maintaining beta cell mass and preventing apoptosis in islets; an induction of angiogenesis, leading to higher glucose uptake by myocytes; a reduction in the oxidative stress-induced insulin resistance and consumption of free fatty acids as metabolic substrate, reducing adipokine generation and consequent inflammation [17,140]. Muscular exercise may also modulate mitochondrial activity, beneficial to glucose homeostasis, and induce changes in the transcription factors involved in insulin signal transduction, which, in turn, increase insulin sensitivity [140,141,142].
IR is a strong determinant of chronic diseases in adulthood, and childhood and adolescence are vulnerable times for the development of IR and its consequences; therapeutical strategies such as intensive lifestyle modification (including improved dietary intake and muscular exercises) undertaken during or prior to puberty may have great positive effects on health outcomes, both in terms of glucose homeostasis (preventing IR and T2D) and in terms of weight control (reducing overweight and preventing obesity development) [143].
Reasonable fears related to the potential for disordered eating and eating disorders upon strict dietary modification in this population led to a larger emphasis on exercise training with respect to nutritional interventions during childhood and adolescence [144]. Indeed, physical activity has become a fundamental of obesity-induced IR treatment [145].
In general, the exercise training types evaluated in children and adolescents can be subdivided into three major categories: aerobic training (AT), resistance training (RT), and combined training (AT + RT), as shown in Table 2.
Kazeminasab et al. performed a meta-analysis to determine the effects of exercise training (AT, RT, RT+RT) on IR and body weight in children and adolescents with overweight or obesity [17]. The authors reviewed 35 studies involving 1,550 children and adolescents aged 9 to 18 years with overweight and obesity, analyzing the effects of exercise training on fasting glucose, fasting insulin, HOMA-IR, and body weight [17]. The interventions lasted between 4 and 24 weeks, with most studies focusing on a 12-week duration. Training sessions were typically held 2 to 5 times per week, with 3 sessions per week being the most common. Each session lasted between 30 and 60 min [17]. The meta-analysis concluded that exercise is an effective strategy to decrease fasting glucose, fasting insulin, HOMA-IR, and BW in children and adolescents who are overweight or obese, and it could be considered an important strategy to control IR [17]. Moreover, the study evidenced that exercise training was effective regardless of the length of the intervention (> 8 and ≤8 weeks), biological sex (female/male), BMI percentiles (overweight/obesity), and health status (with and without other diagnoses). It is worth underscoring that the outcomes evidence were obtained with no specific caloric restriction and/or adherence to a dietary plan; this is important as adherence to a restrictive diet may be challenging for this population [17].
Interestingly, subgroup analyses by sex indicated a significant reduction in fasting glucose, fasting insulin, and HOMA-IR for females and males but not for females and males combined, compared with a control group [17].
Analyzing the type of exercise performed, the authors evidenced that significant reductions in fasting glucose, fasting insulin, and HOMA-IR were obtained both for AT and for CT, but not for RT alone, compared with controls [17]. Exercise training in children with overweight and obesity was also shown to effectively reduce body weight in these patients [17,146].
The results confirmed the data previously obtained by another systematic review and meta-analysis performed by Marson et al., who assessed the associations of aerobic, resistance, and combined exercise with changes in IR, fasting glucose, and fasting insulin in children and adolescents with overweight and obesity. Indeed, the authors showed that physical training, in general, was associated with reductions in fasting insulin levels and HOMA-IR and concluded that exercise training, especially aerobic, leading to a reduction in fasting insulin levels and HOMA-IR, may prevent metabolic syndrome and type 2 diabetes in children and adolescents with obesity and overweight [14]. Moreover, the reduction in fasting glucose and insulin upon exercise was relevant regardless of the length of the interventions. Indeed, significant results were obtained both in the case of long-term interventions (≥8 weeks) and short-term ones (<8 weeks) [17]. Differently from insulin and glucose blood levels, the reduction in body weight was significant for intervention durations of more than 8 weeks but not for durations of less than 8 weeks, and subgroup analyses by sex evidenced a significant reduction in body weight for males but not for females, compared with a control group [17].
Among aerobic training, Kim et al. included jumping rope, which was shown to decrease fasting insulin, fasting blood glucose, and HOMA-IR [147]. Moreover, this aerobic training type was also associated with a decrease in body weight and blood pressure levels [147]. Similarly, Murphy et al. evaluated dancing (in the form of exergaming) and reported beneficial effects in terms of blood pressure levels and lipid profile [148]. Vasconcellos et al. studied the effects of 12-week soccer training (60 min sessions performed 3 times/week) in terms of biochemical, cardiovascular, and fitness health markers in obese people with obesity and evidenced significant reductions in terms of BMI, waist circumference, and blood pressure levels [149]. Moreover, a better lipid blood profile (reduction in total cholesterol and triglycerides) and a decrease in insulin resistance (evaluated with HOMA-IR) were demonstrated [149]. Among resistance training, Seo et al. evaluated the effect of an 8-week yoga-asana training on body composition, lipid profile, and IR in adolescent boys with obesity [150]. The authors showed that the proposed training was effective in weight loss, improvement of body composition, and amelioration of lipid profile and IR, suggesting that yoga may be effective in controlling some metabolic syndrome factors in adolescents with obesity [150].
It remains not fully elucidated whether combined AT and RT may have greater improvements with respect to aerobic exercise alone [17] and whether high-intensity training intervention (HIIT) has more beneficial effects compared with moderate ones [17]. Precisely, HIIT is defined as an alternation of short bursts of intense physical activity, performed with a “near maximal” effort, with brief periods of rest or lower-intensity exercise [151]. Cao et al. showed not significant differences in terms of blood glucose, HOMA-IR, and body weight when comparing HIIT with moderate-intensity ones in children and adolescents [152], but HIIT intervention was shown to improve cardiorespiratory fitness more significantly than moderate-intensity training by Liu et al. [153].
Recently, Garcìa-Hermoso et al. performed a systematic review and network meta-analysis evaluating exercise training types in children and adolescents with a mean age of 13.5 years with excess weight in order to compare training modalities and their association with changes in IR markers [154]. Their study included works with a total of 3051 children and adolescents, 50.4% girls and 49.6% boys, and the authors confirmed that exercise was associated with reductions in FI and HOMA-IR and that a combination of HIIT and resistance training was the most effective training modalities to ameliorate IR markers [154]. Notably, the authors found a nonlinear association between exercise dose and IR markers (IR and HOMA-IR), with a minimal dosage requirement of around 900–1200 MET-min/week, which can be considered equivalent to two to three sessions per week of 1 h of moderate/vigorous training [154]. Importantly, the review also suggests that even a modest increase in weekly exercise can improve glucidic metabolism [154].
Table 2. Studies evaluating the metabolic effects of exercise (AT, RT, and combined) in overweight and/or children and adolescents with obesity.
Table 2. Studies evaluating the metabolic effects of exercise (AT, RT, and combined) in overweight and/or children and adolescents with obesity.
ReferencesExercise TypeAge
(yrs)
Duration (Weeks)DietResults
Glucidic
Metabolism
Anthropometric Parameters:Others
Aerobic training
Sun et al., 2011
[155]
AerobicExe (F): 13.80 ± 0.60
Exe (M): 13.38 ± 0.40
Con: 13.60 ± 0.70
10Habitual dietNo significant effect on FI and HOMA-IRDecrease in BWna
Kelly et al., 2004
[156]
AerobicExe: 10.90 ± 1.99
Con: 11.00 ± 2.24
8Habitual dietDecrease in FBG and FIDecrease in BWAmelioration of lipid profiles
Liu et al., 2018
[157]
AerobicExe: 14.60 ± 0.70
Con: 14.70 ± 0.80
41400–1600 kcal/dayDecrease in FBG, FI, and HOMA-IRDecrease in BWna
Karacabey et al., 2009
[158]
AerobicExe: 11.80 ± 0.50
Con: 11.20 ± 0.80
12Specific diet programDecrease in FIDecrease in BWDecrease in cortisol levels and amelioration of lipid profile
McCormack et al., 2014
[159]
AerobicExe:13.80 ± 2.20
Con:12.10 ± 1.20
8Habitual dietDecrease in HOMA-IRDecrease in BW and WCAmelioration in body composition
Kim et al., 2011
[160]
AerobicExe: 17.63 ± 0.49
Con na
12Habitual dietDecrease in FBG,
FI and
HOMA-IR
Decrease in BWAmelioration of lipid profile
Leite et al., 2022
[161]
AerobicExe and Con: 13.00 ± 1.9012Nutritional guidance in Con, Habitual diet in ExDecrease in FI, FBG,
HOMA-IR and
QUICKI
Decrease in BW and BMIAmelioration of lipid profile
Salahshoornezhad et al., 2022
[162]
AerobicExe and Con: 10.50 ± 1.0210naDecrease in FBGDecrease in BW and WCAmelioration of lipid profile
Alizadeh et al., 2019
[163]
AerobicExe and Con: 18.00 ± 1.56Habitual dietDecrease in FBG
FI and
HOMA-IR
Decrease in BWna
Kim et al., 2020
[147]
AerobicExe and Con 15.00 ± 1.0012Habitual dietDecrease in FBG
FI and
HOMA-IR
Decrease in BWDecrease in blood pressure levels
Murphy et al., 2009
[148]
AerobicExe and Con: 10.21 ± 1.6712Habitual dietnaDecrease in BWDecrease in blood pressure levels and lipid profile
Vasconcellos et al., 2015
[149]
AerobicExe1: 16.60 ± 0.90
Exe2: 16.50 ± 1.20
Con: 16.90 ± 1.00
12Habitual dietDecrease in
HOMA-IR
Decrease in BW and WCDecrease in fat mass, blood pressure, and amelioration of lipid levels
Boer et al., 2014
[164]
AerobicExe1: 18.00 ± 3.20
Exe2: 16.70 ± 3.60
Con: 17.40 ± 2.40
15Habitual dietDecrease in FBG, FI, and HOMA-IRDecrease in BWDecrease in body fat,
blood pressure levels and amelioration of lipid profile
Resistance training
Benson et al., 2008[165]
ResistanceExe and Con: 12.20 ± 1.38Habitual dietDecrease in FBG,
FI
Decrease in WCDecrease in body fat
Kelly et al., 2019
[166]
ResistanceExe: 15.29 ± 0.95
Con: 15.58 ± 0.99
16Habitual dietDecrease in FBGnana
Lee et al., 2013
[167]
Aerobic vs. ResistanceExe1: 14.60 ± 1.90
Exe2: 14.80 ± 1.90
Con: 15.00 ± 2.20
12Habitual dietDecrease in
FI and HOMA-IR
naDecrease in visceral fat
Lee et al., 2010
[168]
Aerobic vs. ResistanceExe1: 15.20 ± 1.90
Exe2: 14.60 ± 1.50
Con: 14.80 ± 1.40
12Habitual dietDecrease in FIDecrease in BWna
Rasooli et al., 2021
[169]
ResistanceExe and Con: 14.00–17.008Habitual dietDecrease in
FI and FBG
Decrease in BWna
Seo et al., 2012
[150]
ResistanceExe: 14.70 ± 1.51
Con: 14.60 ± 3.03
8Balanced diet
Amelioration of IRDecrease in BWAmelioration of lipid profile
Combined training
Chae et al., 2010
[170]
Combined (Aerobic and Resistance)Exe: 10.60 ± 3.80
Con: 10.40 ± 3.10
12<1800–2000 kcal/daynaDecrease in BWImprovement of body composition and serum lipid profiles
Vissers et al., 2008
[171]
Combined (Aerobic and Resista ce)Exe (F): 17.50 ± 1.30
Exe (M): 18.10 ± 1.30
Con (F): 17.10 ± 1.10
Con (M): 17.50 ± 1.40
24Counseling by a dietitianDecrease in
FBG
Decrease in BW and WCna
Wong et al., 2018
[172]
Combined (Aerobic and Resistance)Exe: 15.20 ± 1.20
Con: 15.30 ± 1.10
12Balanced dietDecrease in
FI and FBG
Decrease in BWDecrease in body fat
Sefat et al., 2019
[173]
Combined (Aerobic and Resistance)Exe: 12.40 ± 1.71
Con: 11.80 ± 2.20
8Habitual dietDecrease in
FI, FBG, and HOMA-IR
Decrease in BWna
Son et al., 2017
[174]
Combined (Aerobic and Resistance)Exe and Con: 15.00 ± 4.4712Habitual dietDecrease in
FI, FBG, and HOMA-IR
Decrease in BW and WCna
Zehsaz et al., 2016
[175]
Combined (Aerobic and Resistance)Exe: 10.80 ± 0.90
Con: 10.30 ± 0.90
16Habitual dietDecrease in
FI and HOMA-IR
Decrease in BW and WCDecrease in body fat
and amelioration of lipid profiles
Davis et al., 2011
[176]
Combined (Aerobic and Resistance)Exe: 15.70 ± 1.10
Con: 15.80 ± 1.00
16Habitual dietDecrease in
FI and HOMA-IR
Decrease in WCDecrease in body fat
Farpour-Lambert et al., 2009
[177]
Combined (Aerobic and Resistance)Exe: 9.10 ± 1.40
Con 1: 8.80 ± 1.60
Con 2: 8.50 ± 1.50
12Habitual dietnaDecrease in BWDecrease in
blood pressure levels
Lopes et al., 2016
[178]
Combined (Aerobic and Resistance)Exe: 14.60 ± 1.15
Con: 14.40 ± 1.16
12Habitual dietDecrease in FBG, FI, and HOMA-IRDecrease in BWna
Davis et al., 2009
[179]
Resistance Combined (Aerobic and Resistance)Exe1: 15.70 ± 1.20
Exe2: 14.80 ± 1.00
Con: 15.3 ±0 1.10
16Nutritional education classNo significant changes in FBG and FIDecrease in BWDecrease in body fat
de Lira et al., 2017
[180]
HIIT
LIT
Exe1: 14.95 ± 1.35
Exe2: 14.77 ± 0.94
Con: 14.72 ± 1.35
12Balanced diet
No significant changes in FBG
FI
HOMA-IR
naImprovement of biomarkers related to non-alcoholic fatty liver disease
Meng et al., 2022
[181]
HIIT
MICT
Exe1: 11.4 ± 0.80
Exe2: 11.2 ± 0.70
Con: 11.0 ± 0.70
12Habitual dietDecrease in
FI and
HOMA-IR
Decrease in BMIna
Plavsic et al., 2020
[182]
HIITExe: 16.20 ± 1.30
Con: 15.50 ± 1.50
121500–1700 kcal/dayNo significant changes in metabolic parametersDecrease in BWna
Abassi et al., 2020
[183]
HIIT
MIIT
Exe1: 16.10 ± 0.99
Exe2: 16.50 ± 1.07
Con: 16.90 ± 1.64
12Habitual dietDecrease in FI and
HOMA-IR
Decrease in BWDecrease in body fat
Racil et al., 2016
[184]
HIIT Combined (Plyometric exercise and HIIT)Exe1: 16.60 ± 0.90
Exe2: 16.50 ± 1.20
Con: 16.90 ± 1.00
12Habitual dietDecrease in FBG, FI, and HOMA-IRDecrease in BWDecrease in blood pressure levels
Dias et al., 2018
[185]
HIIT MICTExe1: 12.40 ± 1.90
Exe2: 11.90 ± 2.40
Con: 11.8 ± 2.40
12Balanced dietNo significant reductions in metabolic parametersnaNo significant reductions in metabolic parameters
Racil et al., 2013
[186]
HIIT MIITExe1: 15.60 ± 0.70
Exe2: 16.30 ± 0.52
Con: 15.90 ± 1.20
12Habitual dietDecrease in FI and HOMA-IRDecrease in BWAmelioration of lipid profile
Meyer et al., 2006
[187]
Swimming and Aqua aerobic training
+ Sports games +
Walking
(combined)
Exe and Con: 14.70 ± 2.2024Habitual dietDecrease in FI and
HOMA-IR
naDecrease in body fat, blood pressure levels, CV risk, and amelioration of lipid profile
Others
De Souza et al., 2022
[188]
Karate trainingExe and Con: 12.00–17.0012Nutritional planDecrease in FBGnaAmelioration of lipid profile and heart rate
WC: waist circumference; Con; control; Exe: exercise; FBG: fasting blood glucose, FI: fasting insulin, HOMA-IR: Homeostasis model assessment: insulin resistance, BW: body weight, IR: insulin resistance, Exe: exercise group, Con: controls, HIIT: High-Intensity Interval Training, MICT: moderate-intensity continuous training, MIIT: moderate-intensity interval training, LIIT: Low-Intensity Interval Training, na: not available; CV: cardiovascular; QUICKI: quantitative insulin sensitivity check index.

5. Limits

We recognize that this review has some limitations. Firstly, we present a non-systematic overview and analysis of the existing literature on a particular topic. The non-systematic nature of narrative reviews [18] implies that there are no formally established guidelines for their conduct, potentially introducing selection biases of the literature and often leading to qualitative rather than quantitative syntheses. For instance, our review only considered articles available on PubMed and Scopus, meaning that relevant studies indexed in other databases or search engines might have been inadvertently excluded.
Secondly, in most studies describing the impact of physical exercise on glucose metabolism and IR in the pediatric age, the variations of myokines during training or after a specific exercise time program are not reported. Therefore, it is difficult to qualitatively and quantitatively assess their different roles in various types of activities and to drive more specific conclusions about their particular role in this context.
Additionally, longitudinal studies on long-term follow-up are limited. Further studies are necessary to better evaluate the differences among exercise modalities in terms of outcomes and long-term follow-up.
Finally, the reported studies do not examine in detail the effects of exercise during the transition from childhood to adolescence and from adolescence to adulthood, periods in which both muscle structure and metabolism undergo significant physiological changes. Comprehensive and multidisciplinary studies are necessary to fully understand the pathogenetic mechanisms underlying the association between skeletal muscle health and metabolic balance during growth.

6. Conclusions

Skeletal muscle is recognized as an endocrine organ. The analysis of various molecules produced and secreted by skeletal muscle reveals that the skeletal muscle secretome, through its paracrine and endocrine functions, significantly contributes to the maintenance and regulation of overall physiological health. Specifically, myokines such as IL-6, IL-8, and IL-15, as well as irisin, myonectin, and myostatin, appear to play a crucial role in IR.
Unfortunately, evidences concerning the role of other exercise-induced molecules in glucidic metabolism in the pediatric field are still limited thus, other studies are needed. Additionally, in children with obesity, skeletal muscle can also become a target of obesity-induced and IR-induced inflammation. In the correlation between muscle, IR, and inflammation, the role of infiltration by immune cells and the microvasculature as the site of insulin action and delivery may also be considered.
In the pediatric population, exercise training, regardless of the type of exercise chosen, was shown to have beneficial effects on glycemic metabolism—evaluated with FBG, FI, and HOMA-IR—as well as on BW. This is extremely important to prevent IR, T2D, and the deleterious consequences of these conditions during childhood and adolescence. Physical activity should be recommended as adjunctive therapy to control IR and related outcomes in these patients. It is still unknown whether the effect of exercise training would be similar in both biological sexes, across different ranges of ages (young children vs. adolescents), and whether they would be dependent on overweight or obesity status or other health conditions. Thus, further investigations should focus on these factors to determine the best recommendations.
In addition, it remains unclear which exercise approach is the best; however, aerobic exercise is highly effective in improving insulin sensitivity in children and adolescents with obesity. However, when combined with RT, specifically with HIIT, the benefits seem to be amplified, making this combination the most effective strategy for managing IR. High-intensity activities offer superior metabolic advantages and are often more enjoyable for children, which increases the likelihood of long-term adherence. Therefore, it is crucial to encourage daily participation in these activities, with a strong emphasis on making exercise fun and engaging to ensure consistent, long-term commitment and effective management of glucose metabolism.
Even though the choice of sport should be made considering the child’s physical and cognitive abilities, as well as the coordination skills that can be developed at different ages, at every age, physical activity can be recommended. It is essential to educate people from childhood to develop the habit of engaging in daily physical activity so that it becomes a part of their lifestyle as they grow older. Through education in movement, the developmental process is enhanced and completed, contributing to achieving balanced growth and development and skeletal muscle health while preventing diseases in later stages of life.
The development of strategies and actions to promote physical activity in children and adolescents must be a top priority for public health, not only in terms of individual quality of life and well-being but also for the health of the community, contributing to the reduction in direct and indirect healthcare costs.

Author Contributions

Conceptualization: V.C., M.V., L.M. and G.Z.; independent collection of the contributions: V.C.M., A.B., V.R. and A.G.; writing—original draft preparation, V.C., V.C.M., A.B., V.R., A.G., M.V. and L.M.; writing—review and editing, V.C., V.C.M., A.B., V.R., A.G., M.V. and L.M.; supervision, V.C., M.V., L.M. and G.Z. All authors have read and agreed to the published version of the manuscript.

Funding

Project performed in the context of (1) National Recovery and Resilience Plan (NRRP), Mission 4 Component 2 Investment 1.3-Call for proposal No. 341 of 15 March 2022 of the Italian Ministry of University and Research funded by the European Union-NextGenerationEU. Project code PE00000003, Concession Decree No. 1550 of 11 October 2022 adopted by the Italian Ministry of University and Research, CUP D93C22000890001, Project title “ON Foods—Research and innovation network on food and nutrition Sustainability, Safety, and Security—Working ON Foods”; (2) PODiaCar Project 101128946—PODiaCar—EU4H-2022-PJ-3 co-funded by the European Union.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kirk, B.; Feehan, J.; Lombardi, G.; Duque, G. Muscle, Bone, and Fat Crosstalk: The Biological Role of Myokines, Osteokines, and Adipokines. Curr. Osteoporos. Rep. 2020, 18, 388–400. [Google Scholar] [CrossRef] [PubMed]
  2. Wolfe, R.R. The Underappreciated Role of Muscle in Health and Disease. Am. J. Clin. Nutr. 2006, 84, 475–482. [Google Scholar] [CrossRef]
  3. Lombardi, G. Exercise-Dependent Modulation of Bone Metabolism and Bone Endocrine Function: New Findings and Therapeutic Perspectives. J. Sci. Sport Exerc. 2019, 1, 20–28. [Google Scholar] [CrossRef]
  4. Kim, S.; Choi, J.-Y.; Moon, S.; Park, D.-H.; Kwak, H.-B.; Kang, J.-H. Roles of Myokines in Exercise-Induced Improvement of Neuropsychiatric Function. Pflug. Arch. 2019, 471, 491–505. [Google Scholar] [CrossRef] [PubMed]
  5. Calcaterra, V.; Verduci, E.; Vandoni, M.; Rossi, V.; Fiore, G.; Massini, G.; Berardo, C.; Gatti, A.; Baldassarre, P.; Bianchi, A.; et al. The Effect of Healthy Lifestyle Strategies on the Management of Insulin Resistance in Children and Adolescents with Obesity: A Narrative Review. Nutrients 2022, 14, 4692. [Google Scholar] [CrossRef]
  6. Brandt, C.; Pedersen, B.K. The Role of Exercise-Induced Myokines in Muscle Homeostasis and the Defense against Chronic Diseases. J. Biomed. Biotechnol. 2010, 2010, 520258. [Google Scholar] [CrossRef] [PubMed]
  7. Petersen, A.M.W.; Pedersen, B.K. The Anti-Inflammatory Effect of Exercise. J. Appl. Physiol. (1985) 2005, 98, 1154–1162. [Google Scholar] [CrossRef] [PubMed]
  8. Merz, K.E.; Thurmond, D.C. Role of Skeletal Muscle in Insulin Resistance and Glucose Uptake. In Comprehensive Physiology; Terjung, R., Ed.; Wiley: Hoboken, NJ, USA, 2020; pp. 785–809. ISBN 978-0-470-65071-4. [Google Scholar]
  9. Severinsen, M.C.K.; Pedersen, B.K. Muscle–Organ Crosstalk: The Emerging Roles of Myokines. Endocr. Rev. 2020, 41, 594–609. [Google Scholar] [CrossRef]
  10. Raschke, S.; Eckardt, K.; Bjørklund Holven, K.; Jensen, J.; Eckel, J. Identification and Validation of Novel Contraction-Regulated Myokines Released from Primary Human Skeletal Muscle Cells. PLoS ONE 2013, 8, e62008. [Google Scholar] [CrossRef]
  11. Weiss, R.; Kaufman, F.R. Metabolic Complications of Childhood Obesity: Identifying and Mitigating the Risk. Diabetes Care 2008, 31 (Suppl. 2), S310–S316. [Google Scholar] [CrossRef]
  12. Chang, C.; Liu, W.; Zhao, X.; Li, S.; Yu, C. Effect of Supervised Exercise Intervention on Metabolic Risk Factors and Physical Fitness in Chinese Obese Children in Early Puberty. Obes. Rev. 2008, 9 (Suppl. 1), 135–141. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, G.W.; Lin, J.E.; Blomain, E.S.; Waldman, S.A. Antiobesity Pharmacotherapy: New Drugs and Emerging Targets. Clin. Pharmacol. Ther. 2013, 95, 53–66. [Google Scholar] [CrossRef]
  14. Marson, E.C.; Delevatti, R.S.; Prado, A.K.G.; Netto, N.; Kruel, L.F.M. Effects of Aerobic, Resistance, and Combined Exercise Training on Insulin Resistance Markers in Overweight or Obese Children and Adolescents: A Systematic Review and Meta-Analysis. Prev. Med. 2016, 93, 211–218. [Google Scholar] [CrossRef]
  15. Ross, M.; Kargl, C.K.; Ferguson, R.; Gavin, T.P.; Hellsten, Y. Exercise-Induced Skeletal Muscle Angiogenesis: Impact of Age, Sex, Angiocrines and Cellular Mediators. Eur. J. Appl. Physiol. 2023, 123, 1415–1432. [Google Scholar] [CrossRef] [PubMed]
  16. Islam, H.; Gillen, J.B. Skeletal Muscle Mechanisms Contributing to Improved Glycemic Control Following Intense Interval Exercise and Training. Sports Med. Health Sci. 2023, 5, 20–28. [Google Scholar] [CrossRef] [PubMed]
  17. Kazeminasab, F.; Sharafifard, F.; Miraghajani, M.; Behzadnejad, N.; Rosenkranz, S.K. The Effects of Exercise Training on Insulin Resistance in Children and Adolescents with Overweight or Obesity: A Systematic Review and Meta-Analysis. Front. Endocrinol. (Lausanne) 2023, 14, 1178376. [Google Scholar] [CrossRef]
  18. Gregory, A.T.; Denniss, A.R. An Introduction to Writing Narrative and Systematic Reviews—Tasks, Tips and Traps for Aspiring Authors. Heart Lung Circ. 2018, 27, 893–898. [Google Scholar] [CrossRef]
  19. Goldstein, M.S. Humoral Nature of the Hypoglycemic Factor of Muscular Work. Diabetes 1961, 10, 232–234. [Google Scholar] [CrossRef]
  20. Pedersen, B.K.; Steensberg, A.; Fischer, C.; Keller, C.; Keller, P.; Plomgaard, P.; Febbraio, M.; Saltin, B. Searching for the Exercise Factor: Is IL-6 a Candidate? J. Muscle Res. Cell Motil. 2003, 24, 113–119. [Google Scholar] [CrossRef]
  21. Febbraio, M.A.; Hiscock, N.; Sacchetti, M.; Fischer, C.P.; Pedersen, B.K. Interleukin-6 Is a Novel Factor Mediating Glucose Homeostasis during Skeletal Muscle Contraction. Diabetes 2004, 53, 1643–1648. [Google Scholar] [CrossRef]
  22. Pedersen, B.K.; Febbraio, M.A. Muscles, Exercise and Obesity: Skeletal Muscle as a Secretory Organ. Nat. Rev. Endocrinol. 2012, 8, 457–465. [Google Scholar] [CrossRef] [PubMed]
  23. Hoffmann, C.; Weigert, C. Skeletal Muscle as an Endocrine Organ: The Role of Myokines in Exercise Adaptations. Cold Spring Harb Perspect Med. 2017, 7, a029793. [Google Scholar] [CrossRef] [PubMed]
  24. Hartwig, S.; Raschke, S.; Knebel, B.; Scheler, M.; Irmler, M.; Passlack, W.; Muller, S.; Hanisch, F.-G.; Franz, T.; Li, X.; et al. Secretome Profiling of Primary Human Skeletal Muscle Cells. Biochim. Biophys. Acta 2014, 1844, 1011–1017. [Google Scholar] [CrossRef] [PubMed]
  25. Le Bihan, M.-C.; Bigot, A.; Jensen, S.S.; Dennis, J.L.; Rogowska-Wrzesinska, A.; Lainé, J.; Gache, V.; Furling, D.; Jensen, O.N.; Voit, T.; et al. In-Depth Analysis of the Secretome Identifies Three Major Independent Secretory Pathways in Differentiating Human Myoblasts. J. Proteom. 2012, 77, 344–356. [Google Scholar] [CrossRef] [PubMed]
  26. Bouzakri, K.; Plomgaard, P.; Berney, T.; Donath, M.Y.; Pedersen, B.K.; Halban, P.A. Bimodal Effect on Pancreatic β-Cells of Secretory Products from Normal or Insulin-Resistant Human Skeletal Muscle. Diabetes 2011, 60, 1111–1121. [Google Scholar] [CrossRef]
  27. Hittel, D.S.; Berggren, J.R.; Shearer, J.; Boyle, K.; Houmard, J.A. Increased Secretion and Expression of Myostatin in Skeletal Muscle from Extremely Obese Women. Diabetes 2009, 58, 30–38. [Google Scholar] [CrossRef]
  28. Pratesi, A.; Tarantini, F.; Di Bari, M. Skeletal Muscle: An Endocrine Organ. Clin. Cases Min. Bone Metab. 2013, 10, 11–14. [Google Scholar] [CrossRef]
  29. McPherron, A.C.; Lawler, A.M.; Lee, S.J. Regulation of Skeletal Muscle Mass in Mice by a New TGF-Beta Superfamily Member. Nature 1997, 387, 83–90. [Google Scholar] [CrossRef]
  30. Palsgaard, J.; Brøns, C.; Friedrichsen, M.; Dominguez, H.; Jensen, M.; Storgaard, H.; Spohr, C.; Torp-Pedersen, C.; Borup, R.; De Meyts, P.; et al. Gene Expression in Skeletal Muscle Biopsies from People with Type 2 Diabetes and Relatives: Differential Regulation of Insulin Signaling Pathways. PLoS ONE 2009, 4, e6575. [Google Scholar] [CrossRef]
  31. Schuelke, M.; Wagner, K.R.; Stolz, L.E.; Hübner, C.; Riebel, T.; Kömen, W.; Braun, T.; Tobin, J.F.; Lee, S.-J. Myostatin Mutation Associated with Gross Muscle Hypertrophy in a Child. N. Engl. J. Med. 2004, 350, 2682–2688. [Google Scholar] [CrossRef]
  32. Hamrick, M.W.; Arounleut, P.; Kellum, E.; Cain, M.; Immel, D.; Liang, L.-F. Recombinant Myostatin (GDF-8) Propeptide Enhances the Repair and Regeneration of Both Muscle and Bone in a Model of Deep Penetrant Musculoskeletal Injury. J. Trauma. 2010, 69, 579–583. [Google Scholar] [CrossRef] [PubMed]
  33. Zhu, J.; Li, Y.; Shen, W.; Qiao, C.; Ambrosio, F.; Lavasani, M.; Nozaki, M.; Branca, M.F.; Huard, J. Relationships between Transforming Growth Factor-Β1, Myostatin, and Decorin. J. Biol. Chem. 2007, 282, 25852–25863. [Google Scholar] [CrossRef] [PubMed]
  34. Rue, N.; Vissing, J.; Galbo, H. Insulin Resistance and Increased Muscle Cytokine Levels in Patients with Mitochondrial Myopathy. J. Clin. Endocrinol. Metab. 2014, 99, 3757–3765. [Google Scholar] [CrossRef]
  35. Paulsen, G.; Mikkelsen, U.R.; Raastad, T.; Peake, J.M. Leucocytes, Cytokines and Satellite Cells: What Role Do They Play in Muscle Damage and Regeneration Following Eccentric Exercise? Exerc. Immunol. Rev. 2012, 18, 42–97. [Google Scholar]
  36. Toth, K.G.; McKay, B.R.; De Lisio, M.; Little, J.P.; Tarnopolsky, M.A.; Parise, G. IL-6 Induced STAT3 Signalling Is Associated with the Proliferation of Human Muscle Satellite Cells Following Acute Muscle Damage. PLoS ONE 2011, 6, e17392. [Google Scholar] [CrossRef] [PubMed]
  37. Serrano, A.L.; Baeza-Raja, B.; Perdiguero, E.; Jardí, M.; Muñoz-Cánoves, P. Interleukin-6 Is an Essential Regulator of Satellite Cell-Mediated Skeletal Muscle Hypertrophy. Cell Metab. 2008, 7, 33–44. [Google Scholar] [CrossRef]
  38. Petersen, E.W.; Carey, A.L.; Sacchetti, M.; Steinberg, G.R.; Macaulay, S.L.; Febbraio, M.A.; Pedersen, B.K. Acute IL-6 Treatment Increases Fatty Acid Turnover in Elderly Humans in Vivo and in Tissue Culture in Vitro. Am. J. Physiol. Endocrinol. Metab. 2005, 288, E155–E162. [Google Scholar] [CrossRef]
  39. Ellingsgaard, H.; Ehses, J.A.; Hammar, E.B.; Van Lommel, L.; Quintens, R.; Martens, G.; Kerr-Conte, J.; Pattou, F.; Berney, T.; Pipeleers, D.; et al. Interleukin-6 Regulates Pancreatic Alpha-Cell Mass Expansion. Proc. Natl. Acad. Sci. USA 2008, 105, 13163–13168. [Google Scholar] [CrossRef]
  40. Ellingsgaard, H.; Hauselmann, I.; Schuler, B.; Habib, A.M.; Baggio, L.L.; Meier, D.T.; Eppler, E.; Bouzakri, K.; Wueest, S.; Muller, Y.D.; et al. Interleukin-6 Enhances Insulin Secretion by Increasing Glucagon-like Peptide-1 Secretion from L Cells and Alpha Cells. Nat. Med. 2011, 17, 1481–1489. [Google Scholar] [CrossRef]
  41. Mauer, J.; Chaurasia, B.; Goldau, J.; Vogt, M.C.; Ruud, J.; Nguyen, K.D.; Theurich, S.; Hausen, A.C.; Schmitz, J.; Brönneke, H.S.; et al. Signaling by IL-6 Promotes Alternative Activation of Macrophages to Limit Endotoxemia and Obesity-Associated Resistance to Insulin. Nat. Immunol. 2014, 15, 423–430. [Google Scholar] [CrossRef]
  42. Pedersen, L.; Idorn, M.; Olofsson, G.H.; Lauenborg, B.; Nookaew, I.; Hansen, R.H.; Johannesen, H.H.; Becker, J.C.; Pedersen, K.S.; Dethlefsen, C.; et al. Voluntary Running Suppresses Tumor Growth through Epinephrine- and IL-6-Dependent NK Cell Mobilization and Redistribution. Cell Metab. 2016, 23, 554–562. [Google Scholar] [CrossRef] [PubMed]
  43. Li, Y.; Li, J.; Zhu, J.; Sun, B.; Branca, M.; Tang, Y.; Foster, W.; Xiao, X.; Huard, J. Decorin Gene Transfer Promotes Muscle Cell Differentiation and Muscle Regeneration. Mol. Ther. 2007, 15, 1616–1622. [Google Scholar] [CrossRef]
  44. Broholm, C.; Laye, M.J.; Brandt, C.; Vadalasetty, R.; Pilegaard, H.; Pedersen, B.K.; Scheele, C. LIF Is a Contraction-Induced Myokine Stimulating Human Myocyte Proliferation. J. Appl. Physiol. (1985) 2011, 111, 251–259. [Google Scholar] [CrossRef] [PubMed]
  45. Görgens, S.W.; Hjorth, M.; Eckardt, K.; Wichert, S.; Norheim, F.; Holen, T.; Lee, S.; Langleite, T.; Birkeland, K.I.; Stadheim, H.K.; et al. The Exercise-Regulated Myokine Chitinase-3-like Protein 1 Stimulates Human Myocyte Proliferation. Acta Physiol. (Oxf.) 2016, 216, 330–345. [Google Scholar] [CrossRef]
  46. Kanzleiter, T.; Rath, M.; Görgens, S.W.; Jensen, J.; Tangen, D.S.; Kolnes, A.J.; Kolnes, K.J.; Lee, S.; Eckel, J.; Schürmann, A.; et al. The Myokine Decorin Is Regulated by Contraction and Involved in Muscle Hypertrophy. Biochem. Biophys. Res. Commun. 2014, 450, 1089–1094. [Google Scholar] [CrossRef]
  47. Heinrich, P.C.; Behrmann, I.; Müller-Newen, G.; Schaper, F.; Graeve, L. Interleukin-6-Type Cytokine Signalling through the Gp130/Jak/STAT Pathway. Biochem. J. 1998, 334 Pt 2, 297–314. [Google Scholar] [CrossRef]
  48. Metcalf, D. The Unsolved Enigmas of Leukemia Inhibitory Factor. Stem. Cells 2003, 21, 5–14. [Google Scholar] [CrossRef] [PubMed]
  49. Kwak, S.-Y.; Seo, I.H.; Chung, I.; Kim, S.A.; Lee, J.O.; Lee, H.J.; Kim, S.E.; Han, J.A.; Kang, M.J.; Kim, S.J.; et al. Effect of Chitinase-3-like Protein 1 on Glucose Metabolism: In Vitro Skeletal Muscle and Human Genetic Association Study. FASEB J. 2020, 34, 13445–13460. [Google Scholar] [CrossRef]
  50. Görgens, S.W.; Eckardt, K.; Elsen, M.; Tennagels, N.; Eckel, J. Chitinase-3-like Protein 1 Protects Skeletal Muscle from TNFα-Induced Inflammation and Insulin Resistance. Biochem. J. 2014, 459, 479–488. [Google Scholar] [CrossRef]
  51. Dijk, W.; Beigneux, A.P.; Larsson, M.; Bensadoun, A.; Young, S.G.; Kersten, S. Angiopoietin-like 4 Promotes Intracellular Degradation of Lipoprotein Lipase in Adipocytes. J. Lipid Res. 2016, 57, 1670–1683. [Google Scholar] [CrossRef]
  52. Gray, N.E.; Lam, L.N.; Yang, K.; Zhou, A.Y.; Koliwad, S.; Wang, J.-C. Angiopoietin-like 4 (Angptl4) Protein Is a Physiological Mediator of Intracellular Lipolysis in Murine Adipocytes. J. Biol. Chem. 2012, 287, 8444–8456. [Google Scholar] [CrossRef] [PubMed]
  53. Ben-Zvi, D.; Barrandon, O.; Hadley, S.; Blum, B.; Peterson, Q.P.; Melton, D.A. Angptl4 Links α-Cell Proliferation Following Glucagon Receptor Inhibition with Adipose Tissue Triglyceride Metabolism. Proc. Natl. Acad. Sci. USA 2015, 112, 15498–15503. [Google Scholar] [CrossRef] [PubMed]
  54. Son, Y.; Paton, C.M. A Review of Free Fatty Acid-Induced Cell Signaling, Angiopoietin-like Protein 4, and Skeletal Muscle Differentiation. Front. Physiol. 2022, 13, 987977. [Google Scholar] [CrossRef] [PubMed]
  55. Li, G.; Zhang, H.; Ryan, A.S. Skeletal Muscle Angiopoietin-Like Protein 4 and Glucose Metabolism in Older Adults after Exercise and Weight Loss. Metabolites 2020, 10, 354. [Google Scholar] [CrossRef] [PubMed]
  56. van der Kolk, B.W.; Goossens, G.H.; Jocken, J.W.; Kersten, S.; Blaak, E.E. Angiopoietin-Like Protein 4 and Postprandial Skeletal Muscle Lipid Metabolism in Overweight and Obese Prediabetics. J. Clin. Endocrinol. Metab. 2016, 101, 2332–2339. [Google Scholar] [CrossRef]
  57. Enoki, Y.; Nagai, T.; Hamamura, Y.; Osa, S.; Nakamura, H.; Taguchi, K.; Watanabe, H.; Maruyama, T.; Matsumoto, K. The G Protein-Coupled Receptor Ligand Apelin-13 Ameliorates Skeletal Muscle Atrophy Induced by Chronic Kidney Disease. J. Cachexia Sarcopenia Muscle 2023, 14, 553–564. [Google Scholar] [CrossRef]
  58. Dray, C.; Knauf, C.; Daviaud, D.; Waget, A.; Boucher, J.; Buléon, M.; Cani, P.D.; Attané, C.; Guigné, C.; Carpéné, C.; et al. Apelin Stimulates Glucose Utilization in Normal and Obese Insulin-Resistant Mice. Cell Metab. 2008, 8, 437–445. [Google Scholar] [CrossRef]
  59. Attané, C.; Foussal, C.; Le Gonidec, S.; Benani, A.; Daviaud, D.; Wanecq, E.; Guzmán-Ruiz, R.; Dray, C.; Bezaire, V.; Rancoule, C.; et al. Apelin Treatment Increases Complete Fatty Acid Oxidation, Mitochondrial Oxidative Capacity, and Biogenesis in Muscle of Insulin-Resistant Mice. Diabetes 2012, 61, 310–320. [Google Scholar] [CrossRef]
  60. Hu, G.; Wang, Z.; Zhang, R.; Sun, W.; Chen, X. The Role of Apelin/Apelin Receptor in Energy Metabolism and Water Homeostasis: A Comprehensive Narrative Review. Front. Physiol. 2021, 12, 632886. [Google Scholar] [CrossRef]
  61. Le Moal, E.; Liu, Y.; Collerette-Tremblay, J.; Dumontier, S.; Fabre, P.; Molina, T.; Dort, J.; Orfi, Z.; Denault, N.; Boutin, J.; et al. Apelin Stimulation of the Vascular Skeletal Muscle Stem Cell Niche Enhances Endogenous Repair in Dystrophic Mice. Sci. Transl. Med. 2024, 16, eabn8529. [Google Scholar] [CrossRef]
  62. 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] [PubMed]
  63. Boström, P.; Wu, J.; Jedrychowski, M.P.; Korde, A.; Ye, L.; Lo, J.C.; Rasbach, K.A.; Boström, 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. Nature 2012, 481, 463–468. [Google Scholar] [CrossRef] [PubMed]
  64. Wu, Y.; Wu, Y.; Yu, J.; Zhang, Y.; Li, Y.; Fu, R.; Sun, Y.; Zhao, K.; Xiao, Q. Irisin Ameliorates D-Galactose-Induced Skeletal Muscle Fibrosis via the PI3K/Akt Pathway. Eur. J. Pharmacol. 2023, 939, 175476. [Google Scholar] [CrossRef] [PubMed]
  65. Hoier, B.; Hellsten, Y. Exercise-Induced Capillary Growth in Human Skeletal Muscle and the Dynamics of VEGF. Microcirculation 2014, 21, 301–314. [Google Scholar] [CrossRef] [PubMed]
  66. Andrzejewski, W.; Kassolik, K.; Kobierzycki, C.; Grzegrzolka, J.; Ratajczak-Wielgomas, K.; Jablonska, K.; Halski, T.; Dziegiel, P.; Gworys, B.; Podhorska-Okolow, M. Increased Skeletal Muscle Expression of VEGF Induced by Massage and Exercise. Folia. Histochem. Cytobiol. 2015, 53, 145–151. [Google Scholar] [CrossRef]
  67. Kivelä, R.; Kyröläinen, H.; Selänne, H.; Komi, P.V.; Kainulainen, H.; Vihko, V. A Single Bout of Exercise with High Mechanical Loading Induces the Expression of Cyr61/CCN1 and CTGF/CCN2 in Human Skeletal Muscle. J. Appl. Physiol. 2007, 103, 1395–1401. [Google Scholar] [CrossRef]
  68. Laske, C.; Stransky, E.; Leyhe, T.; Eschweiler, G.W.; Wittorf, A.; Richartz, E.; Bartels, M.; Buchkremer, G.; Schott, K. Stage-Dependent BDNF Serum Concentrations in Alzheimer’s Disease. J. Neural. Transm. (Vienna) 2006, 113, 1217–1224. [Google Scholar] [CrossRef]
  69. Matthews, V.B.; Aström, M.-B.; Chan, M.H.S.; Bruce, C.R.; Krabbe, K.S.; Prelovsek, O.; Akerström, T.; Yfanti, C.; Broholm, C.; Mortensen, O.H.; et al. Brain-Derived Neurotrophic Factor Is Produced by Skeletal Muscle Cells in Response to Contraction and Enhances Fat Oxidation via Activation of AMP-Activated Protein Kinase. Diabetologia 2009, 52, 1409–1418. [Google Scholar] [CrossRef]
  70. Wahren, J. GLUCOSE TURNOVER DURING EXERCISE IN MAN*. Ann. N. Y. Acad. Sci. 1977, 301, 45–55. [Google Scholar] [CrossRef]
  71. Das, A.M.; Steuerwald, U.; Illsinger, S. Inborn Errors of Energy Metabolism Associated with Myopathies. J. Biomed. Biotechnol. 2010, 2010, 340849. [Google Scholar] [CrossRef]
  72. Westerblad, H.; Bruton, J.D.; Katz, A. Skeletal Muscle: Energy Metabolism, Fiber Types, Fatigue and Adaptability. Exp. Cell Res. 2010, 316, 3093–3099. [Google Scholar] [CrossRef] [PubMed]
  73. DeFronzo, R.A.; Tripathy, D. Skeletal Muscle Insulin Resistance Is the Primary Defect in Type 2 Diabetes. Diabetes Care 2009, 32, S157. [Google Scholar] [CrossRef] [PubMed]
  74. Mueckler, M. Family of Glucose-Transporter Genes: Implications for Glucose Homeostasis and Diabetes. Diabetes 1990, 39, 6–11. [Google Scholar] [CrossRef] [PubMed]
  75. Klip, A.; Volchuk, A.; He, L.; Tsakiridis, T. The Glucose Transporters of Skeletal Muscle. Semin. Cell Dev. Biol. 1996, 7, 229–237. [Google Scholar] [CrossRef]
  76. Evans, P.L.; McMillin, S.L.; Weyrauch, L.A.; Witczak, C.A. Regulation of Skeletal Muscle Glucose Transport and Glucose Metabolism by Exercise Training. Nutrients 2019, 11, 2432. [Google Scholar] [CrossRef]
  77. Zisman, A.; Peroni, O.D.; Abel, E.D.; Michael, M.D.; Mauvais-Jarvis, F.; Lowell, B.B.; Wojtaszewski, J.F.P.; Hirshman, M.F.; Virkamaki, A.; Goodyear, L.J.; et al. Targeted Disruption of the Glucose Transporter 4 Selectively in Muscle Causes Insulin Resistance and Glucose Intolerance. Nat. Med. 2000, 6, 924–928. [Google Scholar] [CrossRef]
  78. Ryder, J.W.; Yang, J.; Galuska, D.; Rincón, J.; Björnholm, M.; Krook, A.; Lund, S.; Pedersen, O.; Wallberg-Henriksson, H.; Zierath, J.R.; et al. Use of a Novel Impermeable Biotinylated Photolabeling Reagent to Assess Insulin- and Hypoxia-Stimulated Cell Surface GLUT4 Content in Skeletal Muscle from Type 2 Diabetic Patients. Diabetes 2000, 49, 647–654. [Google Scholar] [CrossRef] [PubMed]
  79. Hp, L. In Vivo Imaging of GLUT4 Translocation. Appl. Physiol. Nutr. Metab. = Physiol. Appl. Nutr. Metab. 2009, 34, 420–423. [Google Scholar] [CrossRef]
  80. Klip, A.; McGraw, T.E.; James, D.E. Thirty Sweet Years of GLUT4. J. Biol. Chem. 2019, 294, 11369–11381. [Google Scholar] [CrossRef]
  81. Jensen, T.E.; Sylow, L.; Rose, A.J.; Madsen, A.B.; Angin, Y.; Maarbjerg, S.J.; Richter, E.A. Contraction-Stimulated Glucose Transport in Muscle Is Controlled by AMPK and Mechanical Stress but Not Sarcoplasmatic Reticulum Ca2+ Release. Mol. Metab. 2014, 3, 742. [Google Scholar] [CrossRef]
  82. Murray, B.; Rosenbloom, C. Fundamentals of Glycogen Metabolism for Coaches and Athletes. Nutr. Rev. 2018, 76, 243. [Google Scholar] [CrossRef] [PubMed]
  83. Paneque, A.; Fortus, H.; Zheng, J.; Werlen, G.; Jacinto, E. The Hexosamine Biosynthesis Pathway: Regulation and Function. Genes 2023, 14, 933. [Google Scholar] [CrossRef]
  84. Wu, C.-L.; Satomi, Y.; Walsh, K. RNA-Seq and Metabolomic Analyses of Akt1-Mediated Muscle Growth Reveals Regulation of Regenerative Pathways and Changes in the Muscle Secretome. BMC Genom. 2017, 18, 181. [Google Scholar] [CrossRef] [PubMed]
  85. Battistuzzi, G.; D’Urso, M.; Toniolo, D.; Persico, G.M.; Luzzatto, L. Tissue-Specific Levels of Human Glucose-6-Phosphate Dehydrogenase Correlate with Methylation of Specific Sites at the 3’ End of the Gene. Proc. Natl. Acad. Sci. USA 1985, 82, 1465–1469. [Google Scholar] [CrossRef] [PubMed]
  86. Abdul-Ghani, M.A.; DeFronzo, R.A. Pathogenesis of Insulin Resistance in Skeletal Muscle. J. Biomed. Biotechnol. 2010, 2010, 476279. [Google Scholar] [CrossRef]
  87. Lee, S.-H.; Park, S.-Y.; Choi, C.S. Insulin Resistance: From Mechanisms to Therapeutic Strategies. Diabetes Metab. J. 2022, 46, 15–37. [Google Scholar] [CrossRef]
  88. Petersen, M.C.; Shulman, G.I. Mechanisms of Insulin Action and Insulin Resistance. Physiol. Rev. 2018, 98, 2133–2223. [Google Scholar] [CrossRef]
  89. DeFronzo, R.A.; Jacot, E.; Jequier, E.; Maeder, E.; Wahren, J.; Felber, J.P. The Effect of Insulin on the Disposal of Intravenous Glucose: Results from Indirect Calorimetry and Hepatic and Femoral Venous Catheterization. Diabetes 1981, 30, 1000–1007. [Google Scholar] [CrossRef]
  90. Steensberg, A.; Van Hall, G.; Osada, T.; Sacchetti, M.; Saltin, B.; Pedersen, B.K. Production of Interleukin-6 in Contracting Human Skeletal Muscles Can Account for the Exercise-induced Increase in Plasma Interleukin-6. J. Physiol. 2000, 529, 237–242. [Google Scholar] [CrossRef]
  91. Kang, S.; Narazaki, M.; Metwally, H.; Kishimoto, T. Historical Overview of the Interleukin-6 Family Cytokine. J. Exp. Med. 2020, 217, e20190347. [Google Scholar] [CrossRef]
  92. Whitham, M.; Chan, M.H.S.; Pal, M.; Matthews, V.B.; Prelovsek, O.; Lunke, S.; El-Osta, A.; Broenneke, H.; Alber, J.; Brüning, J.C.; et al. Contraction-Induced Interleukin-6 Gene Transcription in Skeletal Muscle Is Regulated by c-Jun Terminal Kinase/Activator Protein-1. J. Biol. Chem. 2012, 287, 10771–10779. [Google Scholar] [CrossRef] [PubMed]
  93. Welc, S.S.; Clanton, T.L. The Regulation of Interleukin-6 Implicates Skeletal Muscle as an Integrative Stress Sensor and Endocrine Organ. Exp. Physiol. 2013, 98, 359–371. [Google Scholar] [CrossRef] [PubMed]
  94. Hojman, P.; Brolin, C.; Nørgaard-Christensen, N.; Dethlefsen, C.; Lauenborg, B.; Olsen, C.K.; Åbom, M.M.; Krag, T.; Gehl, J.; Pedersen, B.K. IL-6 Release from Muscles during Exercise Is Stimulated by Lactate-Dependent Protease Activity. Am. J. Physiol.-Endocrinol. Metab. 2019, 316, E940–E947. [Google Scholar] [CrossRef] [PubMed]
  95. Sun, H.; Ma, Y.; Gao, M.; Liu, D. IL-15/sIL-15Rα Gene Transfer Induces Weight Loss and Improves Glucose Homeostasis in Obese Mice. Gene Ther. 2016, 23, 349–356. [Google Scholar] [CrossRef] [PubMed]
  96. Lacraz, G.; Rakotoarivelo, V.; Labbé, S.M.; Vernier, M.; Noll, C.; Mayhue, M.; Stankova, J.; Schwertani, A.; Grenier, G.; Carpentier, A.; et al. Correction: Deficiency of Interleukin-15 Confers Resistance to Obesity by Diminishing Inflammation and Enhancing the Thermogenic Function of Adipose Tissues. PLoS ONE 2016, 11, e0166537. [Google Scholar] [CrossRef]
  97. Yang, H.; Chang, J.; Chen, W.; Zhao, L.; Qu, B.; Tang, C.; Qi, Y.; Zhang, J. Treadmill Exercise Promotes Interleukin 15 Expression in Skeletal Muscle and Interleukin 15 Receptor Alpha Expression in Adipose Tissue of High-Fat Diet Rats. Endocrine 2013, 43, 579–585. [Google Scholar] [CrossRef]
  98. Riechman, S.E.; Balasekaran, G.; Roth, S.M.; Ferrell, R.E. Association of Interleukin-15 Protein and Interleukin-15 Receptor Genetic Variation with Resistance Exercise Training Responses. J. Appl. Physiol. 2004, 97, 2214–2219. [Google Scholar] [CrossRef]
  99. Nielsen, A.R.; Mounier, R.; Plomgaard, P.; Mortensen, O.H.; Penkowa, M.; Speerschneider, T.; Pilegaard, H.; Pedersen, B.K. Expression of Interleukin-15 in Human Skeletal Muscle—Effect of Exercise and Muscle Fibre Type Composition. J. Physiol. 2007, 584, 305–312. [Google Scholar] [CrossRef]
  100. Hingorjo, M.R.; Zehra, S.; Saleem, S.; Qureshi, M.A. Serum Interleukin-15 and Its Relationship with Adiposity Indices before and after Short-Term Endurance Exercise. Pak. J. Med. Sci. 2018, 34, 1125–1131. [Google Scholar] [CrossRef]
  101. Khalafi, M.; Maleki, A.H.; Symonds, M.E.; Sakhaei, M.H.; Rosenkranz, S.K.; Ehsanifar, M.; Korivi, M.; Liu, Y. Interleukin-15 Responses to Acute and Chronic Exercise in Adults: A Systematic Review and Meta-Analysis. Front. Immunol. 2024, 14, 1288537. [Google Scholar] [CrossRef]
  102. Görgens, S.W.; Eckardt, K.; Jensen, J.; Drevon, C.A.; Eckel, J. Exercise and Regulation of Adipokine and Myokine Production. In Progress in Molecular Biology and Translational Science; Elsevier: Amsterdam, The Netherlands, 2015; Volume 135, pp. 313–336. ISBN 978-0-12-803991-5. [Google Scholar]
  103. Li, F.; Li, Y.; Duan, Y.; Hu, C.-A.A.; Tang, Y.; Yin, Y. Myokines and Adipokines: Involvement in the Crosstalk between Skeletal Muscle and Adipose Tissue. Cytokine Growth Factor Rev. 2017, 33, 73–82. [Google Scholar] [CrossRef] [PubMed]
  104. McPherron, A.C.; Lee, S.-J. Double Muscling in Cattle Due to Mutations in the Myostatin Gene. Proc. Natl. Acad. Sci. USA 1997, 94, 12457–12461. [Google Scholar] [CrossRef] [PubMed]
  105. Mosher, D.S.; Quignon, P.; Bustamante, C.D.; Sutter, N.B.; Mellersh, C.S.; Parker, H.G.; Ostrander, E.A. A Mutation in the Myostatin Gene Increases Muscle Mass and Enhances Racing Performance in Heterozygote Dogs. PLoS Genet. 2007, 3, e79. [Google Scholar] [CrossRef] [PubMed]
  106. Kazemi, F. The Correlation of Resistance Exercise-Induced Myostatin with Insulin Resistance and Plasma Cytokines in Healthy Young Men. J. Endocrinol. Investig. 2016, 39, 383–388. [Google Scholar] [CrossRef]
  107. Saremi, A.; Gharakhanloo, R.; Sharghi, S.; Gharaati, M.R.; Larijani, B.; Omidfar, K. Effects of Oral Creatine and Resistance Training on Serum Myostatin and GASP-1. Mol. Cell. Endocrinol. 2010, 317, 25–30. [Google Scholar] [CrossRef]
  108. Louis, E.; Raue, U.; Yang, Y.; Jemiolo, B.; Trappe, S. Time Course of Proteolytic, Cytokine, and Myostatin Gene Expression after Acute Exercise in Human Skeletal Muscle. J. Appl. Physiol. 2007, 103, 1744–1751. [Google Scholar] [CrossRef]
  109. Seldin, M.M.; Wong, G.W. Regulation of Tissue Crosstalk by Skeletal Muscle-Derived Myonectin and Other Myokines. Adipocyte 2012, 1, 200–202. [Google Scholar] [CrossRef]
  110. Seldin, M.M.; Lei, X.; Tan, S.Y.; Stanson, K.P.; Wei, Z.; Wong, G.W. Skeletal Muscle-Derived Myonectin Activates the Mammalian Target of Rapamycin (mTOR) Pathway to Suppress Autophagy in Liver. J. Biol. Chem. 2013, 288, 36073–36082. [Google Scholar] [CrossRef] [PubMed]
  111. Hasegawa, N.; Fujie, S.; Horii, N.; Uchida, M.; Kurihara, T.; Sanada, K.; Hamaoka, T.; Iemitsu, M. Aerobic Exercise Training-Induced Changes in Serum C1q/TNF-Related Protein Levels Are Associated with Reduced Arterial Stiffness in Middle-Aged and Older Adults. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2018, 314, R94–R101. [Google Scholar] [CrossRef]
  112. Park, S.-Y.; Choi, J.H.; Ryu, H.S.; Pak, Y.K.; Park, K.S.; Lee, H.K.; Lee, W. C1q Tumor Necrosis Factor α-Related Protein Isoform 5 Is Increased in Mitochondrial DNA-Depleted Myocytes and Activates AMP-Activated Protein Kinase. J. Biol. Chem. 2009, 284, 27780–27789. [Google Scholar] [CrossRef]
  113. Safdar, A.; Saleem, A.; Tarnopolsky, M.A. The Potential of Endurance Exercise-Derived Exosomes to Treat Metabolic Diseases. Nat. Rev. Endocrinol. 2016, 12, 504–517. [Google Scholar] [CrossRef] [PubMed]
  114. Modan, M.; Halkin, H.; Almog, S.; Lusky, A.; Eshkol, A.; Shefi, M.; Shitrit, A.; Fuchs, Z. Hyperinsulinemia. A Link between Hypertension Obesity and Glucose Intolerance. J. Clin. Investig. 1985, 75, 809–817. [Google Scholar] [CrossRef] [PubMed]
  115. Hardy, O.T.; Czech, M.P.; Corvera, S. What Causes the Insulin Resistance Underlying Obesity? Curr. Opin. Endocrinol. Diabetes Obes. 2012, 19, 81–87. [Google Scholar] [CrossRef] [PubMed]
  116. Li, Y.; Ding, L.; Hassan, W.; Abdelkader, D.; Shang, J. Adipokines and Hepatic Insulin Resistance. J. Diabetes Res. 2013, 2013, 170532. [Google Scholar] [CrossRef]
  117. Maury, E.; Ehala-Aleksejev, K.; Guiot, Y.; Detry, R.; Vandenhooft, A.; Brichard, S.M. Adipokines Oversecreted by Omental Adipose Tissue in Human Obesity. Am. J. Physiol.-Endocrinol. Metab. 2007, 293, E656–E665. [Google Scholar] [CrossRef]
  118. Rabe, K.; Lehrke, M.; Parhofer, K.G.; Broedl, U.C. Adipokines and Insulin Resistance. Mol. Med. 2008, 14, 741–751. [Google Scholar] [CrossRef]
  119. Khan, I.M.; Perrard, X.Y.; Brunner, G.; Lui, H.; Sparks, L.M.; Smith, S.R.; Wang, X.; Shi, Z.-Z.; Lewis, D.E.; Wu, H.; et al. Intermuscular and Perimuscular Fat Expansion in Obesity Correlates with Skeletal Muscle T Cell and Macrophage Infiltration and Insulin Resistance. Int. J. Obes (Lond) 2015, 39, 1607–1618. [Google Scholar] [CrossRef] [PubMed]
  120. Fink, L.N.; Costford, S.R.; Lee, Y.S.; Jensen, T.E.; Bilan, P.J.; Oberbach, A.; Blüher, M.; Olefsky, J.M.; Sams, A.; Klip, A. Pro-Inflammatory Macrophages Increase in Skeletal Muscle of High fat-Fed Mice and Correlate with Metabolic Risk Markers in Humans. Obesity 2014, 22, 747–757. [Google Scholar] [CrossRef]
  121. Liu, J.; Liu, Z. Muscle Insulin Resistance and the Inflamed Microvasculature: Fire from Within. Int. J. Mol. Sci. 2019, 20, 562. [Google Scholar] [CrossRef]
  122. Vincent, M.A.; Dawson, D.; Clark, A.D.H.; Lindner, J.R.; Rattigan, S.; Clark, M.G.; Barrett, E.J. Skeletal Muscle Microvascular Recruitment by Physiological Hyperinsulinemia Precedes Increases in Total Blood Flow. Diabetes 2002, 51, 42–48. [Google Scholar] [CrossRef]
  123. Lebovitz, H.E. Insulin Resistance and Its Treatment by Thiazolidinediones. Recent Prog. Horm. Res. 2001, 56, 265–294. [Google Scholar] [CrossRef] [PubMed]
  124. Todd, M.K.; Watt, M.J.; Le, J.; Hevener, A.L.; Turcotte, L.P. Thiazolidinediones Enhance Skeletal Muscle Triacylglycerol Synthesis While Protecting against Fatty Acid-Induced Inflammation and Insulin Resistance. Am. J. Physiol.-Endocrinol. Metab. 2007, 292, E485–E493. [Google Scholar] [CrossRef]
  125. Sadeghi, H.M.; Adeli, I.; Calina, D.; Docea, A.O.; Mousavi, T.; Daniali, M.; Nikfar, S.; Tsatsakis, A.; Abdollahi, M. Polycystic Ovary Syndrome: A Comprehensive Review of Pathogenesis, Management, and Drug Repurposing. Int. J. Mol. Sci. 2022, 23, 583. [Google Scholar] [CrossRef]
  126. Dabadghao, P. Polycystic Ovary Syndrome in Adolescents. Best Pract. Res. Clin. Endocrinol. Metab. 2019, 33, 101272. [Google Scholar] [CrossRef] [PubMed]
  127. Abdalla, M.A.; Shah, N.; Deshmukh, H.; Sahebkar, A.; Östlundh, L.; Al-Rifai, R.H.; Atkin, S.L.; Sathyapalan, T. The Effect of Thiazolidinediones in Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Adv. Ther. 2024, 41, 2168–2195. [Google Scholar] [CrossRef] [PubMed]
  128. Ehrmann, D.A.; Schneider, D.J.; Sobel, B.E.; Cavaghan, M.K.; Imperial, J.; Rosenfield, R.L.; Polonsky, K.S. Troglitazone Improves Defects in Insulin Action, Insulin Secretion, Ovarian Steroidogenesis, and Fibrinolysis in Women with Polycystic Ovary Syndrome 1. J. Clin. Endocrinol. Metab. 1997, 82, 2108–2116. [Google Scholar] [CrossRef]
  129. Reiser, E.; Lanbach, J.; Böttcher, B.; Toth, B. Non-Hormonal Treatment Options for Regulation of Menstrual Cycle in Adolescents with PCOS. J. Clin. Med. 2022, 12, 67. [Google Scholar] [CrossRef]
  130. Joham, A.E.; Norman, R.J.; Stener-Victorin, E.; Legro, R.S.; Franks, S.; Moran, L.J.; Boyle, J.; Teede, H.J. Polycystic Ovary Syndrome. Lancet Diabetes Endocrinol. 2022, 10, 668–680. [Google Scholar] [CrossRef]
  131. Liu, D.; Li, N.; Zhou, Y.; Wang, M.; Song, P.; Yuan, C.; Shi, Q.; Chen, H.; Zhou, K.; Wang, H.; et al. Sex-specific Associations between Skeletal Muscle Mass and Incident Diabetes: A Population-based Cohort Study. Diabetes Obes. Metab. 2024, 26, 820–828. [Google Scholar] [CrossRef]
  132. Ciarambino, T.; Crispino, P.; Guarisco, G.; Giordano, M. Gender Differences in Insulin Resistance: New Knowledge and Perspectives. Curr. Issues Mol. Biol. 2023, 45, 7845–7861. [Google Scholar] [CrossRef]
  133. Lee, J.M. Insulin Resistance in Children and Adolescents. Rev. Endocr. Metab. Disord. 2007, 7, 141–147. [Google Scholar] [CrossRef] [PubMed]
  134. Wu, H.; Ballantyne, C.M. Metabolic Inflammation and Insulin Resistance in Obesity. Circ. Res. 2020, 126, 1549–1564. [Google Scholar] [CrossRef] [PubMed]
  135. Kojta, I.; Chacińska, M.; Błachnio-Zabielska, A. Obesity, Bioactive Lipids, and Adipose Tissue Inflammation in Insulin Resistance. Nutrients 2020, 12, 1305. [Google Scholar] [CrossRef] [PubMed]
  136. Way, K.L.; Hackett, D.A.; Baker, M.K.; Johnson, N.A. The Effect of Regular Exercise on Insulin Sensitivity in Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Diabetes Metab. J. 2016, 40, 253. [Google Scholar] [CrossRef]
  137. Wahren, J.; Felig, P.; Ahlborg, G.; Jorfeldt, L. Glucose Metabolism during Leg Exercise in Man. J. Clin. Investig. 1971, 50, 2715–2725. [Google Scholar] [CrossRef]
  138. Soo, J.; Raman, A.; Lawler, N.G.; Goods, P.S.R.; Deldicque, L.; Girard, O.; Fairchild, T.J. The Role of Exercise and Hypoxia on Glucose Transport and Regulation. Eur. J. Appl. Physiol. 2023, 123, 1147–1165. [Google Scholar] [CrossRef]
  139. Wallberg-Henriksson, H. Repeated Exercise Regulates Glucose Transport Capacity in Skeletal Muscle. Acta Physiol. Scand. 1986, 127, 39–44. [Google Scholar] [CrossRef]
  140. Yaribeygi, H.; Atkin, S.L.; Simental-Mendía, L.E.; Sahebkar, A. Molecular Mechanisms by Which Aerobic Exercise Induces Insulin Sensitivity. J. Cell. Physiol. 2019, 234, 12385–12392. [Google Scholar] [CrossRef] [PubMed]
  141. Hood, D.A.; Uguccioni, G.; Vainshtein, A.; D’souza, D. Mechanisms of Exercise-Induced Mitochondrial Biogenesis in Skeletal Muscle: Implications for Health and Disease. In Comprehensive Physiology; Prakash, Y.S., Ed.; Wiley: Hoboken, NJ, USA, 2011; pp. 1119–1134. ISBN 978-0-470-65071-4. [Google Scholar]
  142. Masi, L.N.; Serdan, T.D.A.; Levada-Pires, A.C.; Hatanaka, E.; Silveira, L.D.R.; Cury-Boaventura, M.F.; Pithon-Curi, T.C.; Curi, R.; Gorjão, R.; Hirabara, S.M. Regulation of Gene Expression by Exercise-Related Micrornas. Cell. Physiol. Biochem. 2016, 39, 2381–2397. [Google Scholar] [CrossRef]
  143. Watts, K.; Jones, T.W.; Davis, E.A.; Green, D. Exercise Training in Obese Children and Adolescents: Current Concepts. Sports Med. 2005, 35, 375–392. [Google Scholar] [CrossRef]
  144. Stabouli, S.; Erdine, S.; Suurorg, L.; Jankauskienė, A.; Lurbe, E. Obesity and Eating Disorders in Children and Adolescents: The Bidirectional Link. Nutrients 2021, 13, 4321. [Google Scholar] [CrossRef] [PubMed]
  145. West, S.L.; Banks, L.; Schneiderman, J.E.; Caterini, J.E.; Stephens, S.; White, G.; Dogra, S.; Wells, G.D. Physical Activity for Children with Chronic Disease; a Narrative Review and Practical Applications. BMC Pediatr. 2019, 19, 12. [Google Scholar] [CrossRef] [PubMed]
  146. Stoner, L.; Rowlands, D.; Morrison, A.; Credeur, D.; Hamlin, M.; Gaffney, K.; Lambrick, D.; Matheson, A. Efficacy of Exercise Intervention for Weight Loss in Overweight and Obese Adolescents: Meta-Analysis and Implications. Sports Med. 2016, 46, 1737–1751. [Google Scholar] [CrossRef] [PubMed]
  147. Kim, J.; Son, W.-M.; Headid Iii, R.J.; Pekas, E.J.; Noble, J.M.; Park, S.-Y. The Effects of a 12-Week Jump Rope Exercise Program on Body Composition, Insulin Sensitivity, and Academic Self-Efficacy in Obese Adolescent Girls. J. Pediatr. Endocrinol. Metab. 2020, 33, 129–137. [Google Scholar] [CrossRef] [PubMed]
  148. Murphy, E.C.-S.; Carson, L.; Neal, W.; Baylis, C.; Donley, D.; Yeater, R. Effects of an Exercise Intervention Using Dance Dance Revolution on Endothelial Function and Other Risk Factors in Overweight Children. Int. J. Pediatr. Obes. 2009, 4, 205–214. [Google Scholar] [CrossRef] [PubMed]
  149. Vasconcellos, F.; Seabra, A.; Cunha, F.; Montenegro, R.; Penha, J.; Bouskela, E.; Nogueira Neto, J.F.; Collett-Solberg, P.; Farinatti, P. Health Markers in Obese Adolescents Improved by a 12-Week Recreational Soccer Program: A Randomised Controlled Trial. J. Sports Sci. 2016, 34, 564–575. [Google Scholar] [CrossRef]
  150. Seo, D.Y.; Lee, S.; Figueroa, A.; Kim, H.K.; Baek, Y.H.; Kwak, Y.S.; Kim, N.; Choi, T.H.; Rhee, B.D.; Ko, K.S.; et al. Yoga Training Improves Metabolic Parameters in Obese Boys. Korean J. Physiol. Pharmacol. 2012, 16, 175. [Google Scholar] [CrossRef]
  151. Atakan, M.M.; Li, Y.; Koşar, Ş.N.; Turnagöl, H.H.; Yan, X. Evidence-Based Effects of High-Intensity Interval Training on Exercise Capacity and Health: A Review with Historical Perspective. Int. J. Environ. Res. Public Health 2021, 18, 7201. [Google Scholar] [CrossRef]
  152. Cao, M.; Tang, Y.; Li, S.; Zou, Y. Effects of High-Intensity Interval Training and Moderate-Intensity Continuous Training on Cardiometabolic Risk Factors in Overweight and Obesity Children and Adolescents: A Meta-Analysis of Randomized Controlled Trials. Int. J. Environ. Res. Public Health 2021, 18, 11905. [Google Scholar] [CrossRef]
  153. Liu, J.; Zhu, L.; Su, Y. Comparative Effectiveness of High-Intensity Interval Training and Moderate-Intensity Continuous Training for Cardiometabolic Risk Factors and Cardiorespiratory Fitness in Childhood Obesity: A Meta-Analysis of Randomized Controlled Trials. Front. Physiol. 2020, 11, 214. [Google Scholar] [CrossRef]
  154. García-Hermoso, A.; López-Gil, J.F.; Izquierdo, M.; Ramírez-Vélez, R.; Ezzatvar, Y. Exercise and Insulin Resistance Markers in Children and Adolescents With Excess Weight: A Systematic Review and Network Meta-Analysis. JAMA Pediatr. 2023, 177, 1276. [Google Scholar] [CrossRef] [PubMed]
  155. Sun, M.-X.; Huang, X.-Q.; Yan, Y.; Li, B.-W.; Zhong, W.-J.; Chen, J.-F.; Zhang, Y.-M.; Wang, Z.-Z.; Wang, L.; Shi, X.-C.; et al. One-Hour after-School Exercise Ameliorates Central Adiposity and Lipids in Overweight Chinese Adolescents: A Randomized Controlled Trial. Chin. Med. J. (Engl.) 2011, 124, 323–329. [Google Scholar] [PubMed]
  156. Kelly, A.S.; Wetzsteon, R.J.; Kaiser, D.R.; Steinberger, J.; Bank, A.J.; Dengel, D.R. Inflammation, Insulin, and Endothelial Function in Overweight Children and Adolescents: The Role of Exercise. J. Pediatr. 2004, 145, 731–736. [Google Scholar] [CrossRef] [PubMed]
  157. Liu, M.; Lin, X.; Wang, X. Decrease in Serum Chemerin through Aerobic Exercise plus Dieting and Its Association with Mitigation of Cardio-Metabolic Risk in Obese Female Adolescents. J. Pediatr. Endocrinol. Metab. 2018, 31, 127–135. [Google Scholar] [CrossRef] [PubMed]
  158. Karacabey, K. The Effect of Exercise on Leptin, Insulin, Cortisol and Lipid Profiles in Obese Children. J. Int. Med. Res. 2009, 37, 1472–1478. [Google Scholar] [CrossRef]
  159. McCormack, S.E.; McCarthy, M.A.; Harrington, S.G.; Farilla, L.; Hrovat, M.I.; Systrom, D.M.; Thomas, B.J.; Torriani, M.; McInnis, K.; Grinspoon, S.K.; et al. Effects of Exercise and Lifestyle Modification on Fitness, Insulin Resistance, Skeletal Muscle Oxidative Phosphorylation and Intramyocellular Lipid Content in Obese Children and Adolescents. Pediatr. Obes. 2014, 9, 281–291. [Google Scholar] [CrossRef]
  160. Kim, J.Y.; Kim, E.S.; Jeon, J.Y.; Jekal, Y. Improved Insulin Resistance, Adiponectin and Liver Enzymes without Change in Plasma Vaspin Level after 12 Weeks of Exercise Training among Obese Male Adolescents. Korean J. Obes. 2011, 20, 138. [Google Scholar] [CrossRef]
  161. Leite, N.; Tadiotto, M.C.; Corazza, P.R.P.; De Menezes Junior, F.J.; Carli, M.E.C.; Milano-Gai, G.E.; Lopes, W.A.; Gaya, A.R.; Brand, C.; Mota, J.; et al. Responsiveness on Metabolic Syndrome Criteria and Hepatic Parameters after 12 Weeks and 24 Weeks of Multidisciplinary Intervention in Overweight Adolescents. J. Endocrinol. Investig. 2021, 45, 741–752. [Google Scholar] [CrossRef]
  162. Salahshoornezhad, S.; Sohrabi, Z.; Mani, A.; Abdelbasset, W.K.; Mehrabi, M.; Zare, M.; Mehrabani, S.; Gerami, S.; Haghighat, N.; Akbarzadeh, M.; et al. Effect of a Multi-Disciplinary Program on Anthropometric and Biochemical Parameters in Obese and Overweight Elementary School Girls: A Randomized Clinical Trial. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 1982–1989. [Google Scholar] [CrossRef]
  163. Alizadeh, H.; Safarzade, A. Effect of a 6-Week Running Sprint Interval Training Protocol on Serum Meteorin-like Hormone, Insulin Resistance, and Body Composition in Overweight Adolescents. Med. Sport 2019, 72, 79–88. [Google Scholar] [CrossRef]
  164. Boer, P.-H.; Meeus, M.; Terblanche, E.; Rombaut, L.; Wandele, I.D.; Hermans, L.; Gysel, T.; Ruige, J.; Calders, P. The Influence of Sprint Interval Training on Body Composition, Physical and Metabolic Fitness in Adolescents and Young Adults with Intellectual Disability: A Randomized Controlled Trial. Clin. Rehabil. 2014, 28, 221–231. [Google Scholar] [CrossRef]
  165. Benson, A.C.; Torode, M.E.; Fiatarone Singh, M.A. The Effect of High-Intensity Progressive Resistance Training on Adiposity in Children: A Randomized Controlled Trial. Int. J. Obes 2008, 32, 1016–1027. [Google Scholar] [CrossRef]
  166. Kelly, L.; Holmberg, P.M.; Schroeder, E.T.; Loza, A.; Lin, X.; Moody, A.; Hughes, A.; Gibson, A.-M.; Kirk, A. Effect of Home-Based Strength Training Program on IGF-I, IGFBP-1 and IGFBP-3 in Obese Latino Boys Participating in a 16-Week Randomized Controlled Trial. J. Pediatr. Endocrinol. Metab. 2019, 32, 1121–1129. [Google Scholar] [CrossRef]
  167. Lee, S.; Deldin, A.R.; White, D.; Kim, Y.; Libman, I.; Rivera-Vega, M.; Kuk, J.L.; Sandoval, S.; Boesch, C.; Arslanian, S. Aerobic Exercise but Not Resistance Exercise Reduces Intrahepatic Lipid Content and Visceral Fat and Improves Insulin Sensitivity in Obese Adolescent Girls: A Randomized Controlled Trial. Am. J. Physiol.-Endocrinol. Metab. 2013, 305, E1222–E1229. [Google Scholar] [CrossRef]
  168. Lee, S.; Bacha, F.; Hannon, T.; Kuk, J.L.; Boesch, C.; Arslanian, S. Effects of Aerobic Versus Resistance Exercise Without Caloric Restriction on Abdominal Fat, Intrahepatic Lipid, and Insulin Sensitivity in Obese Adolescent Boys. Diabetes 2012, 61, 2787–2795. [Google Scholar] [CrossRef]
  169. Rasooli, S.A.; Fathi, R.; Golzar, F.A.-K.; Baghersalimi, M. The Effect of Circuit Resistance Training on Plasma Levels of Amino Acids, Alpha-Hydroxybutyrate, Mannose, and Urinary Levels of Glycine Conjugated Adducts in Obese Adolescent Boys. Appl. Physiol. Nutr. Metab. 2021, 46, 561–570. [Google Scholar] [CrossRef]
  170. Chae, H.-W.; Kwon, Y.-N.; Rhie, Y.-J.; Kim, H.-S.; Kim, Y.-S.; Paik, I.-Y.; Suh, S.-H.; Kim, D.-H. Effects of a Structured Exercise Program on Insulin Resistance, Inflammatory Markers and Physical Fitness in Obese Korean Children. J. Pediatr. Endocrinol. Metab. 2010, 23, 1065–1072. [Google Scholar] [CrossRef]
  171. Vissers, D.; De Meulenaere, A.; Vanroy, C.; Vanherle, K.; Van De Sompel, A.; Truijen, S.; Van Gaal, L. Effect of a Multidisciplinary School-Based Lifestyle Intervention on Body Weight and Metabolic Variables in Overweight and Obese Youth. e-SPEN Eur. e-J. Clin. Nutr. Metab. 2008, 3, e196–e202. [Google Scholar] [CrossRef]
  172. Wong, A.; Sanchez-Gonzalez, M.A.; Son, W.-M.; Kwak, Y.-S.; Park, S.-Y. The Effects of a 12-Week Combined Exercise Training Program on Arterial Stiffness, Vasoactive Substances, Inflammatory Markers, Metabolic Profile, and Body Composition in Obese Adolescent Girls. Pediatr. Exerc. Sci. 2018, 30, 480–486. [Google Scholar] [CrossRef]
  173. Mohammadi Sefat, S.; Shabani, R.; Nazari, M. The Effect of Concurrent Aerobic-Resistance Training on Thyroid Hormones, Blood Glucose Hemostasis, and Blood Lipid Indices in Overweight Girls with Hypothyroidism. Horm. Mol. Biol. Clin. Investig. 2019, 40, 20190031. [Google Scholar] [CrossRef]
  174. Son, W.-M.; Sung, K.-D.; Bharath, L.P.; Choi, K.-J.; Park, S.-Y. Combined Exercise Training Reduces Blood Pressure, Arterial Stiffness, and Insulin Resistance in Obese Prehypertensive Adolescent Girls. Clin. Exp. Hypertens. 2017, 39, 546–552. [Google Scholar] [CrossRef]
  175. Zehsaz, F.; Farhangi, N.; Ghahramani, M. The Response of Circulating Omentin-1 Concentration to 16-Week Exercise Training in Male Children with Obesity. Physician Sportsmed. 2016, 44, 355–361. [Google Scholar] [CrossRef]
  176. Davis, J.N.; Gyllenhammer, L.E.; Vanni, A.A.; Meija, M.; Tung, A.; Schroeder, E.T.; Spruijt-Metz, D.; Goran, M.I. Startup Circuit Training Program Reduces Metabolic Risk in Latino Adolescents. Med. Sci. Sports Exerc. 2011, 43, 2195–2203. [Google Scholar] [CrossRef]
  177. Farpour-Lambert, N.J.; Aggoun, Y.; Marchand, L.M.; Martin, X.E.; Herrmann, F.R.; Beghetti, M. Physical Activity Reduces Systemic Blood Pressure and Improves Early Markers of Atherosclerosis in Pre-Pubertal Obese Children. J. Am. Coll. Cardiol. 2009, 54, 2396–2406. [Google Scholar] [CrossRef]
  178. Lopes, W.A.; Leite, N.; Da Silva, L.R.; Brunelli, D.T.; Gáspari, A.F.; Radominski, R.B.; Chacon-Mikahil, M.P.T.; Cavaglieri, C.R. Effects of 12 Weeks of Combined Training without Caloric Restriction on Inflammatory Markers in Overweight Girls. J. Sports Sci. 2016, 34, 1902–1912. [Google Scholar] [CrossRef]
  179. Davis, J.N.; Tung, A.; Chak, S.S.; Ventura, E.E.; Byrd-Williams, C.E.; Alexander, K.E.; Lane, C.J.; Weigensberg, M.J.; Spruijt-Metz, D.; Goran, M.I. Aerobic and Strength Training Reduces Adiposity in Overweight Latina Adolescents. Med. Sci. Sports Exerc. 2009, 41, 1494–1503. [Google Scholar] [CrossRef]
  180. De Lira, C.T.; Dos Santos, M.A.; Gomes, P.P.; Fidelix, Y.L.; Dos Santos, A.C.; Tenório, T.R.; Lofrano-Prado, M.C.; Do Prado, W.L. Aerobic Training Performed at Ventilatory Threshold Improves Liver Enzymes and Lipid Profile Related to Non-Alcoholic Fatty Liver Disease in Adolescents with Obesity. Nutr. Health 2017, 23, 281–288. [Google Scholar] [CrossRef]
  181. Meng, C.; Yucheng, T.; Shu, L.; Yu, Z. Effects of School-Based High-Intensity Interval Training on Body Composition, Cardiorespiratory Fitness and Cardiometabolic Markers in Adolescent Boys with Obesity: A Randomized Controlled Trial. BMC Pediatr. 2022, 22, 112. [Google Scholar] [CrossRef]
  182. Plavsic, L.; Knezevic, O.M.; Sovtic, A.; Minic, P.; Vukovic, R.; Mazibrada, I.; Stanojlovic, O.; Hrncic, D.; Rasic-Markovic, A.; Macut, D. Effects of High-Intensity Interval Training and Nutrition Advice on Cardiometabolic Markers and Aerobic Fitness in Adolescent Girls with Obesity. Appl. Physiol. Nutr. Metab. 2020, 45, 294–300. [Google Scholar] [CrossRef]
  183. Abassi, W.; Ouerghi, N.; Ghouili, H.; Haouami, S.; Bouassida, A. Greater Effects of High-Compared with Moderate-Intensity Interval Training on Thyroid Hormones in Overweight/Obese Adolescent Girls. Horm. Mol. Biol. Clin. Investig. 2020, 41, 20200031. [Google Scholar] [CrossRef]
  184. Racil, G.; Zouhal, H.; Elmontassar, W.; Abderrahmane, A.B.; De Sousa, M.V.; Chamari, K.; Amri, M.; Coquart, J.B. Plyometric Exercise Combined with High-Intensity Interval Training Improves Metabolic Abnormalities in Young Obese Females More so than Interval Training Alone. Appl. Physiol. Nutr. Metab. 2016, 41, 103–109. [Google Scholar] [CrossRef] [PubMed]
  185. Dias, K.A.; Ingul, C.B.; Tjønna, A.E.; Keating, S.E.; Gomersall, S.R.; Follestad, T.; Hosseini, M.S.; Hollekim-Strand, S.M.; Ro, T.B.; Haram, M.; et al. Effect of High-Intensity Interval Training on Fitness, Fat Mass and Cardiometabolic Biomarkers in Children with Obesity: A Randomised Controlled Trial. Sports Med. 2018, 48, 733–746. [Google Scholar] [CrossRef]
  186. Racil, G.; Ben Ounis, O.; Hammouda, O.; Kallel, A.; Zouhal, H.; Chamari, K.; Amri, M. Effects of High vs. Moderate Exercise Intensity during Interval Training on Lipids and Adiponectin Levels in Obese Young Females. Eur. J. Appl. Physiol. 2013, 113, 2531–2540. [Google Scholar] [CrossRef] [PubMed]
  187. Meyer, A.A.; Kundt, G.; Lenschow, U.; Schuff-Werner, P.; Kienast, W. Improvement of Early Vascular Changes and Cardiovascular Risk Factors in Obese Children After a Six-Month Exercise Program. J. Am. Coll. Cardiol. 2006, 48, 1865–1870. [Google Scholar] [CrossRef]
  188. De Souza, F.; Da Silva, L.A.; Ferreira, G.S.; De Souza, M.M.M.; Bobinski, F.; Palandi, J.; Marcon, C.E.M.; Martins, D.F.; Schuelter-Trevisol, F.; Trevisol, D.J. Karate Training Improves Metabolic Health in Overweight and Obese Adolescents: A Randomized Clinical Trial. Pediatr. Exerc. Sci. 2022, 34, 108–118. [Google Scholar] [CrossRef]
Figure 1. Glucose metabolism in skeletal muscle. Legend: Created by Biorender®. ECM: extracellular matrix; ICM: intracellular matrix.
Figure 1. Glucose metabolism in skeletal muscle. Legend: Created by Biorender®. ECM: extracellular matrix; ICM: intracellular matrix.
Life 14 01198 g001
Figure 2. Key myokines involved in muscle contraction and influencing glucose metabolism. Created by BioRender®. FBG = fasting blood glucose; FI = fasting insulin; HOMA-IR = Homeostatic Model Assessment for Insulin Resistance index; BW = body weight; BMI = body mass index; WC = waist circumference; NAFLD = Non-alcoholic fatty liver disease.
Figure 2. Key myokines involved in muscle contraction and influencing glucose metabolism. Created by BioRender®. FBG = fasting blood glucose; FI = fasting insulin; HOMA-IR = Homeostatic Model Assessment for Insulin Resistance index; BW = body weight; BMI = body mass index; WC = waist circumference; NAFLD = Non-alcoholic fatty liver disease.
Life 14 01198 g002
Table 1. Myokines and its main functions.
Table 1. Myokines and its main functions.
MyokinesFunction
Myostatin [22,27,29,30,31,32,33]It increases with physical inactivity.
It is an inhibitor of muscle mass gain and bone healing.
It is involved in metabolic equilibrium and the control of adipose tissue activity and mass.
It is regulated by decorin and follistatin.
IL-6 [4,7,23,28,36,37,39,40,41]It is involved in autocrine and paracrine signaling in skeletal muscle, particularly activating the STAT3 pathway, which is important for hypertrophic muscle growth and myogenesis.
It enhances insulin-stimulated glucose uptake and oxidation, stimulates lipolysis and fat oxidation, promotes pancreatic β-cell expansion, and improves insulin secretion and glycemic control.
IL-6 produced by skeletal muscles during exercise exerts anti-inflammatory effects, suppressing the synthesis of IL-1 and TNF-α and stimulating the production of anti-inflammatory cytokines like IL-1ra and IL-10.
IL-15 [4,28]It is an anabolic factor that stimulates muscle growth: IL-15 signaling is involved in the regulation of muscle fiber composition and contractility.
It is involved in lipid metabolism regulation, reducing lipid accumulation in preadipocytes and decreasing the mass of white adipose tissue.
A negative relationship has been observed between plasma IL-15 levels and total body fat mass, especially trunk fat mass.
Decorin [33,43,46]It plays a role in the regulation of muscle hypertrophy, particularly in response to exercise.
Together with follistatin, it is a myostatin inhibitor.
Leukemia inhibitory factor (LIF) [22,44,47,48]It is a contraction-induced myokine.
It acts in an autocrine and/or paracrine fashion.
It promotes platelet production, the proliferation of hematopoietic cells, bone formation, neural survival and development, and the acute-phase response in liver cells.
It supports satellite cell proliferation.
Chitinase-3-like protein 1 (CHI3L1) [45,49,50]It is produced and secreted by skeletal muscle in response to acute physical exercise.
Through the CHI3L1/PAR-2 signaling pathway, it stimulates myocyte proliferation, which is important for skeletal muscle remodeling in response to training.
It is linked to increased glucose uptake in skeletal muscles both via an AMP-activated protein kinase (AMPK)-dependent mechanism and via the PI3K/AKT pathway.
Angiopoietin-like 4 (ANGPTL4) [51,52,53,54,55,56]It inhibits lipoprotein lipase activity, which reduces lipid accumulation and promotes lipolysis in white adipose tissue.
In non-exercising muscles, ANGPTL4 levels increase in response to elevated plasma FFA through PPAR activation, helping to prevent fat overload and ensuring the supply of fatty acids to active skeletal muscles.
By regulating its expression through PPARs, ANGPTL4 may contribute to the development of insulin resistance.
ANGPTL4 levels can be modified by exercise, such as acute resistance and strength training, as well as by dietary factors, particularly the intake of SFA.
It has been implicated in mice as a mediator of pancreatic β-cell hyperplasia.
Apelin [57,58,59,60,61]It enhances glucose uptake in skeletal muscle.
It increases mitochondrial oxidative capacity in skeletal muscle.
It improves muscle function by enhancing energy production.
It reduces inflammation in skeletal muscle.
It supports muscle stem cell (MuSC) regeneration, especially stimulating skeletal muscle endothelial cells (ECs).
Irisin [22,62,63,64]It is a polypeptide hormone produced by muscles in response to physical exercise.
It promotes the conversion of white adipose tissue into a brown fat-like phenotype.
It exerts antifibrotic effects on the heart, liver, pancreas, and muscle.
VEGF [65,66]It is a primary angiogenic factor that promotes the growth of new blood vessels, including the capillaries within skeletal muscle.
During physical exercise, VEGF levels rise in the muscle interstitium, enhancing angiogenic activity.
It facilitates the development and growth of blood vessels within skeletal muscle tissue, ensuring adequate blood supply to the muscles.
It contributes to the regeneration of tissues, supporting repair and recovery processes in skeletal muscle.
IL-8 [22]It induces angiogenesis.
CYR61 (CCN1) and CTGF (CCN2) [67]They increase after physical activity, especially following mechanical loading.
They regulate the expression of genes involved in angiogenesis and ECM remodeling.
Brain-derived neurotrophic factor (BDNF) [28,68,69]Its levels increase after physical activity.
It plays a critical role in the growth, survival, and maintenance of neurons.
It impacts information processing, learning, and memory.
It influences energy metabolism by enhancing fat oxidation in skeletal muscles through an AMPK-dependent pathway.
It contributes to a decrease in adipose tissue mass.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Calcaterra, V.; Magenes, V.C.; Bianchi, A.; Rossi, V.; Gatti, A.; Marin, L.; Vandoni, M.; Zuccotti, G. How Can Promoting Skeletal Muscle Health and Exercise in Children and Adolescents Prevent Insulin Resistance and Type 2 Diabetes? Life 2024, 14, 1198. https://doi.org/10.3390/life14091198

AMA Style

Calcaterra V, Magenes VC, Bianchi A, Rossi V, Gatti A, Marin L, Vandoni M, Zuccotti G. How Can Promoting Skeletal Muscle Health and Exercise in Children and Adolescents Prevent Insulin Resistance and Type 2 Diabetes? Life. 2024; 14(9):1198. https://doi.org/10.3390/life14091198

Chicago/Turabian Style

Calcaterra, Valeria, Vittoria Carlotta Magenes, Alice Bianchi, Virginia Rossi, Alessandro Gatti, Luca Marin, Matteo Vandoni, and Gianvincenzo Zuccotti. 2024. "How Can Promoting Skeletal Muscle Health and Exercise in Children and Adolescents Prevent Insulin Resistance and Type 2 Diabetes?" Life 14, no. 9: 1198. https://doi.org/10.3390/life14091198

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