Skip to main content

Crosstalk between autophagy and insulin resistance: evidence from different tissues

Abstract

Insulin is a critical hormone that promotes energy storage in various tissues, as well as anabolic functions. Insulin resistance significantly reduces these responses, resulting in pathological conditions, such as obesity and type 2 diabetes mellitus (T2DM). The management of insulin resistance requires better knowledge of its pathophysiological mechanisms to prevent secondary complications, such as cardiovascular diseases (CVDs). Recent evidence regarding the etiological mechanisms behind insulin resistance emphasizes the role of energy imbalance and neurohormonal dysregulation, both of which are closely regulated by autophagy. Autophagy is a conserved process that maintains homeostasis in cells. Accordingly, autophagy abnormalities have been linked to a variety of metabolic disorders, including insulin resistance, T2DM, obesity, and CVDs. Thus, there may be a link between autophagy and insulin resistance. Therefore, the interaction between autophagy and insulin function will be examined in this review, particularly in insulin-responsive tissues, such as adipose tissue, liver, and skeletal muscle.

Introduction

Insulin resistance

Type 2 diabetes mellitus (T2DM) is characterized by pathophysiological abnormalities, including impaired insulin secretion from pancreatic islet β-cells, peripheral insulin resistance, and excessive glucose production by the liver. When tissues such as muscle, fat, and liver fail to react properly to insulin, glucose is not readily eliminated from the blood; as a result, the tissues begin to exhibit insulin resistance, which causes the pancreas to produce more insulin in order for the cells to receive glucose [1, 2]. Additionally, pathophysiological conditions such as hyperglycemia, hypertension, visceral adiposity, dyslipidemia, hyperuricemia, elevated inflammatory markers, endothelial dysfunction, and a prothrombic state are metabolically caused by insulin resistance. If insulin resistance continues to increase, it will lead to T2DM, nonalcoholic fatty liver disease (NAFLD), and metabolic syndrome [2]. Decreased insulin binding to its receptor, receptor phosphorylation, tyrosine kinase activity, phosphorylation of insulin receptor substrates (IRSs), and GLUT4 downregulation are observed in insulin-resistant tissues [3]. Other modifications specific to certain tissues will also occur, including changes in the adipocytes of obese individuals with T2DM, decreased insulin receptor substrate (IRS-1) expression, reduced activity of IRS-1-associated phosphoinositide 3-kinase (PI3K), and IRS-2 becoming the main docking protein for PI3K [4]. Nonetheless, it should be noted that even though protein levels of IRS-1 and IRS-2 are normal, the PI3K function linked with both IRSs is impaired in the skeletal muscle of obese T2DM patients [5].

Insulin resistance in obese individuals is caused by a number of mechanisms. As suggested by several studies, proinflammatory cytokines and endoplasmic stress contribute to insulin resistance by activating serine kinases c-Jun N-terminal kinase (JNK) and I kappa B kinase (IKK-b), which are responsible for increasing the phosphorylation of IRS1 at serine sites (serine 302 pS302 and serine 307 pS307). This, in turn, leads to negative regulation of normal insulin signaling [6]. High levels of adipocyte lipid deposition in obese individuals lead to the production of proinflammatory cytokines including interleukin (IL)-6, IL-1b, tumor necrosis factor-alpha (TNF-α), and resistin, resulting in further activation of JNK and nuclear factor kappa B (NF-κB) pathways [7, 8]. In obesity, an increase in fatty acids and their intracellular metabolites (e.g., diacylglycerols) stimulates protein kinase C (PKC), resulting in the activation of serine/threonine kinases and impaired tyrosine phosphorylation of IRS [9, 10].

As major metabolic organelles, mitochondria play a significant role in the insulin signaling of dependent tissues. Insulin is not only required for mitochondria to function properly, but also serves as the basis for the integrity of the mitochondrial electron transport chain (mETC) as it damps down forkhead box O (FOXO) 11/heme oxygenase (HMOX) 12 and maintains a consistent NAD/NADH ratio in mitochondria [11, 12]. Oxidative stress is shown to cause insulin resistance by impairing insulin signal transduction and dysregulating adipokines [13]. In addition, it contributes to the activation of several serine-threonine kinase pathways namely inhibitor of nuclear factor kappa-B kinase subunit beta (IKKβ)/NF‐κB and JNK. These pathways, in turn, lead to the phosphorylation of IRS proteins and consequent IRS degradation [14]. It has been demonstrated that high levels of free radicals suppress the cell membrane GLUT‐4 localization by impairing insulin signaling [15].

Other associations between oxidative stress and insulin resistance include reduced insulin‐caused relocation of IRS‐1 and phosphatidylinositol (PIP) kinase between microsomes and cytoplasm, decreased protein kinase B (PKB) phosphorylation, serine phosphorylation at the serine 307 of IRS‐1, and lowered GLUT‐4 expression [14, 15]. Moreover, it has been found that β-cell endoplasmic reticulum (ER) stress leads to the disruption of insulin synthesis and sensitivity [16, 17].

Autophagy

Autophagy or “self-eating” is a conserved process essential for sustaining cellular homeostasis. This process allows cells to degrade and remove aggregated or misfolded proteins, damaged organelles, and pathogens [18]. During the autophagy pathway, cellular components are transported to lysosomes to be degraded and recycled. There are three main types of autophagy based on how substrates are delivered to the lysosome: chaperon-mediated autophagy, micro-autophagy, and macroautophagy [19]. Since macroautophagy is the preliminary model of autophagy, similar to previous research, we reserve the term autophagy to refer to this type. The components of autophagy machinery and its basic mechanisms were first explored in Saccharomyces cerevisiae, and subsequently, they were studied in other organisms, particularly in mammalians [20]. The process of autophagy generally involves the following steps: induction, formation of the initial nucleus, development of autophagosomal vesicles, combination with lysosomes, and finally digestion. Most proteins implicated in this process are known as autophagy-related (ATG) proteins [21].

In mammalians, the Unc-51-like kinase 1/2 (ULK1/2) complex consisting of ULK1/2, ATG13, FIP200, and ATG 101 is involved in the induction of autophagy [22]. Under conditions of high nutrients or in the presence of growth factors, the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) phosphorylates and inactivates ULK1/2 and ATG13, resulting in the inhibition of autophagy. During starvation (especially amino acid depletion) or in the presence of rapamycin (a specific inhibitor of mTOR), however, mTORC1 disassociates from ULK1, thereby inducing autophagy [22]. Another energy sensor is AMP-activated protein kinase (AMPK), which activates autophagy when ATP levels drop, either directly by phosphorylating ULK1 or indirectly by inactivating mTORC1 [23]. Active ULK then stimulates class III PI3K or the vacuolar protein sorting 34 (Vps34) complex and recruits it to the phagophore assembly site (PAS) [24]. The Vps34 complex is composed of Vps34, beclin-1, Vps15, ATG14, and activating molecule in beclin-1-regulated autophagy (AMBRA1) [24]. This complex synthesizes phosphatidylinositol triphosphate (PI3P), which is required for phagophore localization. Phagophore is a special membrane that starts the sequestration of cytoplasmic constituents. Elongation of phagophore to a double membrane vesicle (called autophagosome) completes the sequestration [25]. Two conjugation systems promote autophagosome formation: ATG5-ATG12-ATG16L and microtubule-associated protein 1 light chain 3 (LC3). LC3-I is a cleaved form of LC3 which is converted to LC3-II by lipidation as autophagy progresses [19]. A rise in LC3-II levels, according to the increased number of autophagosomes, suggests autophagy induction. LC3 serves as a receptor for adaptor protein p62/sequestosome 1 (SQSTM1), which can interact with ubiquitinated substrates, and make them a target for lysosomes [26]. The autophagy pathway is completed by the fusion of lysosomes and autophagosomes, leading to the formation of autophagolysosomes whose contents are degraded and recycled. P62 itself is degraded in autophagolysosomes and since its level is used as a tool to assess the autophagic flux, an increase in p62 levels denotes autophagy flux inhibition and vice versa [26] (Fig. 1).

Fig. 1
figure 1

The mechanism of the autophagy pathway in mammalian cells. The autophagy process includes four main steps: induction, phagophore formation, phagophore elongation, and fusion with the lysosome. In the first step, different signals, such as starvation, rapamycin, and similar factors, inhibit mTOR and activate the ULK complex, thereby inducing autophagy. Active ULK stimulates the Vps34 complex to produce PI3P, which is needed for phagophore formation. In the following step, the phagophore expands, engulfs cytoplasmic constituents, and finally fuses with the lysosome to degrade their contents

Autophagy is generally considered a defense mechanism against various stresses, such as nutrient deficiencies or caloric restriction (the most well-known), oxidative injury, and depletion of growth factors, and it promotes cell survival. In unfavorable conditions, it provides nutrients for cells and removes toxic molecules, damaged organelles, and infectious agents [19]. In addition to the maintenance of homeostasis, autophagy regulates the maturation, development, and differentiation of various cells, each with a physiological role (e.g., adipocytes in fat metabolism) [27]. Accordingly, the impairment of this process is associated with pathological conditions including cancer, inflammation, and autoimmunity, as well as metabolic, cardiovascular, and neurodegenerative disorders [28, 29]. Furthermore, autophagy gradually subsides with aging, which is attributed to age-associated diseases such as T2DM [30]. One of the main characteristics of T2DM is insulin resistance. In this review, we discuss the role of autophagy in the target tissues of insulin with a focus on its contribution to the pathophysiology of insulin resistance (Fig. 2).

Fig. 2
figure 2

Molecular mechanism of lipophagy. Under nutrient deprivation, lipophagy forms phagophores. Patatin-like phospholipase domain-containing enzyme (PNPLA) interacts with LDs and performs critical functions in the breakdown of LDs. Autophagosomes engulf LDs and fuse with a lysosome to form an autolysosome. Then, lysosomal lipases hydrolyze the neutral lipids of LDs

Adipose tissue

Altered adipose tissue autophagy in obesity

Obesity, characterized by a remarkable expansion of adipose tissue mass, is the leading risk factor for insulin resistance. Based on previous studies, human adipose tissue and adipocytes both contain autophagosomes and express autophagy genes [31]. Ost et al. originally revealed that the autophagosome content and the autophagic flux were enhanced in adipocytes isolated from obese individuals [32]. This observation was confirmed by studies that reported an enhanced expression of autophagic machinery genes in the adipose tissue of obese patients, such as autophagy-related genes (ATG5, ATG7, and ATG12), beclin-1, LC3A and B, LC3-II, and p62. This increased expression was attributed to the enhanced improved function of both the E2 factor (E2F) and FOXO transcription factors [33, 34]. Interestingly, it has been reported that upregulation of autophagy in obese individuals is more pronounced in omental fat tissue than in subcutaneous fat tissue. Consistently, higher expression of autophagy-related genes was shown in the visceral fat depot, which is linked to metabolic and cardiovascular risks [35, 36]. The rates of fat accumulation and hypertrophy of fat cells are significantly associated with the expression of autophagy genes, and autophagy alteration is accompanied by obesity-associated insulin resistance prior to metabolic and cardiovascular dysfunction [31].

In addition, ER stress, inflammation, and hypoxia, which are the processes that are active in adipose tissue during obesity, promote autophagy via the suppression of mTORC1 [37]. On the whole, autophagy could serve as a protective mechanism against increased inflammation associated with obesity or as a compensatory response to the excessive accumulation of nutrients and damaged organelles in hypertrophic adipocytes [38].

Autophagy, insulin signaling, and insulin resistance in adipose tissue

Insulin, an anabolic hormone, is a potent inhibitor of autophagy [39]. It has been demonstrated that insulin can prevent autophagy by mTORC1 activation, resulting in the suppression of FOXO and ULK1 factors [40]. The PI3K–Akt pathway is a crucial constituent of insulin signaling, which contributes to the inhibition of autophagy by insulin. Akt inhibits FOXO1/3 and induces mTORC1 activity, thereby revealing a major association between insulin signaling and autophagy [34, 41]. However, targeted ATG7 deletion causes detrimental differentiation of WAT, and its browning, resulting in improved insulin sensitivity, and glucose use, as well as enhanced β-oxidation of fatty acids [42, 43]. These differentiations and metabolic changes by autophagy inhibition have a critical role in insulin resistance [34]. However, in a study by Cai et al. on mouse models, specific knockout of ATG3 and ATG16L1 in adipocytes caused insulin resistance. Inhibiting autophagy in the adipocytes, in particular, interfered with insulin signaling to Akt in the WAT, liver, and skeletal muscle [44]. As cells that highly infiltrate into WAT during obesity, macrophages can affect insulin sensitivity and glucose homeostasis [45]. Previous studies have demonstrated that the knockout of ATG5 or ATG7 in macrophages interferes with insulin sensitivity and glucose tolerance [46, 47]. Figure 3 illustrates how autophagy is regulated in both lean and obese adipose tissue, as well as how it affects adipocytes (3.1.a and 3.1.b). Based on these findings, it could be concluded that congenital autophagy inhibition impairs adipogenesis and leads to insulin sensitivity, whereas selective inhibition of this pathway in mature adipocytes results in insulin resistance [34].

Fig. 3
figure 3

The insulin signaling under physiological and pathological conditions in various organs. 3.1: autophagy regulation in lean and obese adipose tissue and its effects on adipocytes. In the lean state (3.1.a), the stimulation of mTORC1 by insulin results in to autophagy inhibition. The inhibition of autophagy induces the ‘browning’ phenotype of the adipocytes. In obesity (3.1.b), ER stress, hypoxia and inflammation stimulate insulin resistance, resulting in mTORC1 inhibition and subsequently to induction of autophagy. Autophagy improve adipocyte function through eliminating damaged organelles and misfolded proteins and prohibiting the proinflammatory responses. Furthermore, excessive stimulation of autophagy may increase energy storage of adipocyte and promote cell death. 3.2: in physiological state (3.2.a), following the binding of insulin to its receptor, Akt is activated and leads to the inhibition of glycogen synthase kinase 3 and forkhead box O (FOXO) 1 in liver. Inhibition of glycogen synthase kinase leads to increased glycogen synthase activity, therefore, increased glycogen synthesis. Following the inhibition of FOXO1, the expression of glucose 6 phosphatase and phosphoenol pyruvate carboxykinase genes decreases, as a result, decreases the hepatic glucose production. Also, insulin decreases autophagy by activating Akt and inhibiting FOXO1. Akt increases lipid synthesis in hepatocytes by activating SREBP1c. The increase in lipid content by SREBP1c causes disruption in lipophagy and hepatic steatosis. In insulin resistance state (3.2.b), glycogen synthase kinase is activated and inhibits glycogen synthase. FOXO1 activity is increased, subsequently, gluconeogenesis pathway, lipid synthesis and very-low-density lipoprotein (VLDL) as well as autophagy are activated. SREBP1c increases its activity through ER stress or through IRS-1. 3.3: in the physiological state during starvation (3.3.a), the insulin hormone binding to its receptors causes the signaling adapter IRS-1 to be recruited and activate AMPK. Active AMPK causes mTORC1 reduction and an increase in autophagy. In a pathological state and during hyperglycemia (3.3.b), the binding of insulin to its receptor causes IRS-1 phosphorylation and recruits PI3K in muscles. Then, the PI3K converts PIP2 to PIP3, as a result, PIP3 induces Akt phosphorylation and increases glucose uptake. Also, Akt phosphorylation increases mTORC1 and decreases autophagy. mTORC1; mammalian target of rapamycin complex 1, ER; endoplasmic reticulum, FOXO1; forkhead box O1, SREBP1c; sterol regulatory element-binding transcription factor 1, VLDL; very-low-density lipoprotein, IRS-1; insulin receptor substrate 1, AMPK; AMP-activated protein kinase, IR; insulin receptor, PI3K; phosphatidylinositol-3-kinase, PIP2; phosphatidylinositol 4,5-bisphosphate, PIP3; phosphatidylinositol-3, 4, 5-triphosphate

Autophagy and adipose tissue development and its contribution in insulin resistance

Adipogenesis is a two-step process in which fibroblast-like, multipotent adipose mesenchymal stem cells (ASCs) are converted to mature adipocytes [33]. As the most frequent cells in adipose tissue, adipocytes are involved in storing energy as fat. During obesity, an increase in the size and number of these cells leads to the expansion of adipose tissue, which is closely connected to insulin resistance [48].

It has been demonstrated that autophagy is involved in the development and differentiation of adipose tissue. Meanwhile, the exact regulatory mechanisms of autophagy in adipogenesis are unknown. As a possible mechanism, autophagosomes facilitate the reorganization of cytoplasmic components via the mobilization of membranes within the cell, thereby contributing to adipogenesis [49]. Furthermore, it has been shown that the stability of peroxisome proliferator-activated receptor (PPAR) γ, which is a major regulator of adipogenesis and adipocyte differentiation, is increased by autophagy [50].

It has been revealed that genetic interference with the process affect pathways linked to adipose differentiation. As an example, ATG5 defect in pre-adipocytes, prevents complete adipocyte differentiation and results in the characteristics of adipose tissue brown fat [42, 51, 52]. In this regard, it has been reported that in ATG5−/− mice, mouse embryonic fibroblasts (MEFs) showed reduced adipogenesis. These cells exhibited signs of adipogenesis initiation and triacylglycerol accumulation, but were not able to complete the maturation of adipocytes [51]. It has also been reported that in mice with the adipose specific Atg7 knockout, the amount of BAT is increased. In fact, in lack of autophagy, progenitor cells fail to complete differentiation and so follow default pathway and create BAT. BAT indicators such as peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α), UCP-1, and mitochondrial enzymes are increased, which is associated with weight loss and increases β-oxidation. BAT development in these animals enhanced insulin sensitivity in body, demonstrating adipose tissue autophagy has a large effect on insulin sensitivity [42]. Furthermore, based on in vivo studies, mTORC1 inhibition and consequent autophagy activation in BAT can result in an increase in thermogenesis and the induction of lipolysis. Accordingly, it could be concluded that mTORC1 signaling and autophagy contribute to lipolysis regulation, and consequently, adipocyte development [53].

In addition to the role of autophagy in preadipocyte differentiation, it was supposed that its contribution in adipocyte maturation. In 3T3-L1 cells with ATG5 and ATG7 deficiency, C/EBPβ, which contributes to clonal expansion of preadipocyte and activates the transcription factors for adipocyte maturation, was downregulated [42]. The observed defective adipogenesis in animals with ATG7 deletion in skeletal muscle suggest that changes in autophagic activity can play a role in the modulation of inter-organ crosstalk. Therefore, any primary/developmental disturbance in autophagy can influence the mass and homeostasis of adipose tissue [54].

Accordingly, autophagy inhibition could be considered a preventative measure against insulin resistance and obesity by increasing the development of BAT. However, the molecular mechanisms by which autophagy induces the trans differentiation of white adipose tissue (WAT) to BAT should be identified [48].

Autophagy and mitochondrial dysfunction, their role in insulin resistance

Obesity and excessive nutrient consumption can lead to mitochondrial dysfunctions. The highest levels of mitochondrial dysfunctions in obesity were observed in adipose tissues, liver, and muscles [55]. Mitochondrial dysfunction enhances mitophagy, which is a quality control pathway for selective clearance of the excess mitochondria when mitochondria have accumulated during adipogenesis or have been damaged by oxidative stress [56,57,58]. It has been suggested that changes in the mitochondrial mass and function play a key role in the pathophysiology of insulin resistance [59]. Since in obesity, increased autophagy in adipose tissue results in elevated mitochondrial degradation, it is likely that elevated autophagy reduces the mitochondrial mass in adipocytes, increases the energy storage potential, and contributes to the pathogenesis of obesity [60]. Recent studies on autophagy in adipocytes have focused on the mitochondrial turnover pathways. Mice with adipocyte-specific inhibition of autophagy are resistant to diet-induced obesity and show improved insulin sensitivity [42, 43]. The findings suggest that autophagy has a critical function in adipocyte development through the clearance of mitochondria.

Autophagy and ER stress in adipose tissue and their relationship with insulin resistance

Growing evidence indicates that ER stress and insulin resistance are potentially connected through autophagy [61,62,63]. Similarly, insulin resistance and autophagy-dependent apoptosis are likely to be caused by unresolved ER stress due to the activation of JNK phosphorylation by inositol-requiring kinase 1 (IRE1). Moreover, obesity induces ER stress in adipocytes, resulting in an increase in adiponectin levels, and the insulin receptor is misfolded and subsequently degraded through the autophagy-dependent pathway [62]. Zhou et al. showed that insulin receptors were significantly downregulated in 3T3-L1 adipocytes when were treated with ER stress inducers, as well as in the fat tissues of obese individuals [64]. It appears that autophagy has a role in the downregulation of insulin receptors in a condition of ER stress, such that the autophagy inhibitor 3-methyladenine (3-MA) blocked the ER-mediated degradation of insulin receptors [64]. Nonetheless, the ER stress-induced insulin signaling impairment in 3T3L1 adipocytes was not improved by blocking autophagy, indicating the dysfunction of served insulin receptors in the presence of ER stress [64]. It can be concluded that autophagy is stimulated in response to ER stress to clear toxic aggregates such as impaired IR.

Moreover, it was shown that the peripheral target tissues of insulin are the sites wherein autophagy is triggered by ER stress-induced insulin resistance [65]. On the account that some processes link autophagic disorders with ER stress responses and impaired insulin signaling through activating the JNK/IKK signaling pathways, ER stress, autophagy, and altered adipose tissue metabolism are all shown to be related to the development of insulin resistance [66].

Li et al. found that failed autophagy makes adipocytes susceptible to ER stress, which in turn leads to autophagy and insulin resistance in an IRE1–JNK pathway-dependent manner in mature adipocytes [62]. Overall, the downregulation of autophagy results in the growth of misfolded large molecules and aged/damaged organelles which would, in return, lead to the development of insulin resistance and ER stress. In general, the role of autophagy and its relation with ER stress and insulin resistance in adipose tissue are regarded as double-edged, and further analysis is needed to determine whether this role is protective or detrimental.

Autophagy and adipose tissue inflammation and their effects on insulin resistance

Inflammation is one of the key processes leading to the development of insulin resistance in obesity [67]. It has been shown that the inflamed adipose tissue of obese individuals releases a variety of cytokines, such as IL-18, IL-6, IL-1, and TNF-α, thereby contributing to obesity-induced insulin resistance [38]. Based on previous animal and human studies, in adipose tissue adipocytes and macrophages, autophagy is upregulated as a result of obesity-associated inflammation. This pathway impacts the inflammatory status by regulating the gene expression and secretion of proinflammatory cytokines, such as IL-1, IL-6, and IL-8, which are involved in systemic glucose homeostasis and metabolic diseases. Therefore, it appears that autophagy is the result, not the cause, of obesity-induced inflammation in adipose tissue [38, 68]. In this regard, it has been demonstrated that in obese mice, global ATG7 haplodeficiency leads to insulin resistance as well as the maturation of pro-IL-1β to IL-1β and increased inflammation [69]. Additionally, it has been reported that the deficiency of the autophagic protein ATG16L1 augmented IL-1β processing upon TLR4 stimulation. The exact mechanism by which the gene expression and subsequent secretion of inflammatory cytokines are enhanced after the inhibition of autophagy remains unknown. One possible mechanism may be that the inhibition of autophagy could increase IL-1 production in adipose tissue through the suppression of intracellular pro-IL-1 degradation, triggering the extracellular signal-regulated kinase (ERK)/nuclear factor-B pathway and the activation of inflammasome [70,71,72]. Moreover, mitochondrial autophagy is found to be correlated with insulin resistance and may also regulate the expression of inflammatory genes [73]. As an adipokine, leptin has been reported to decrease autophagy and consequently modulate adipose tissue inflammation. As shown in ob/ob mice, leptin prohibits autophagy through the downregulation of beclin-1 and ATG12, as well as the formation of the activating transcription factor (Atf) 4/ATG5 complex and reduces IĸB degradation by decreasing IL-18 and IL-1β in adipocytes, thus impacting ER stress and inflammation [74].

The role of NLRP3 inflammasome in autophagy-related insulin resistance in adipocytes

Recent studies have shown that NLRP3 inflammasome disrupts metabolic regulation in adipose tissue by activating macrophage infiltration. However, the mechanism of NLRP3 activation and its role in adipocytes remain unclear. In a study by Javaid et al., TNF-α treatment induced NLRP3 expression and caspase-1 activity in adipocytes, partially through dysregulation of autophagy. High-fat diet regulates the expression levels of NLRP3 and caspase-1 in stromal vascular fractions and adipocytes. TNFα induces mitochondrial dysfunction, oxidative stress, and insulin resistance, which activate the adipose NLRP3 inflammasome without altering IL-1β. Active adipocyte NLRP3 inflammasome contributes to mitochondrial dysfunction and insulin resistance. Lipocalin 2, an NLRP3-dependent adipokine, mediated interactions between adipocytes and macrophages. NLRP3 mediates the deleterious effects of TNFα on mitochondria and glucose metabolism and regulates glucose uptake in adipose tissue. Lipocalin 2 from adipocytes could also activate macrophages, suggesting a role for adipose NLRP3 inflammasome activation in fat metabolism. This axis of TNFα–NLRP3–Lcn2 contributes to adipocyte–macrophage interaction and obesity-induced inflammation, creating a vicious cycle in adipose tissue [75].

Targeting autophagy in obesity

Although decreasing caloric intake or absorption and bariatric surgery have been established as the main clinical approaches for the management of obesity, the long-term management of this condition seems to be challenging [76]. Preclinical evidence has proposed using autophagy modulators as a therapeutic approach for the treatment of obesity and metabolic diseases. However, it has not been proven whether the pharmacological targeting of autophagy has clinical efficacy in obesity [77].

In addition to pharmacological interferences, lifestyle modifications, such as exercise and caloric restriction, can counteract obesity and insulin resistance [78, 79]. The findings of previous studies have demonstrated that caloric restriction and exercise enhance autophagy in patients with obesity, and autophagy induction has a critical role in the beneficial impacts of these lifestyle modifications on obesity [80,81,82,83]. It has been shown that drugs that are utilized to reduce caloric intake by appetite inhibition, such as metformin, leptin, adiponectin, the agonists of glucagon-like peptide 1 (GLP1), liraglutide, and exenatide, serve direct regulatory functions in autophagy, however, the modulation of autophagy is mostly related to the inhibition of energy intake [84,85,86,87,88,89]. Moreover, incretin-based therapies, such as treatment with dipeptidyl peptidase 4 (DPP-4) inhibitors, which might impact autophagy, are considered as critical approaches for the treatment of obesity [90]. Based on the findings of animal studies, it has been demonstrated that similar to caloric restriction, ketogenic diets also have metabolic advantages, including resistance to obesity in mice, and increasing autophagy have a role in the beneficial impacts of ketogenic diets [91, 92]. Overall, the role of autophagy in adipose tissue development, and its activation in adipose tissue in obesity, propose that this pathway is an accepted modulator that could be targeted for therapeutic aims, since the inhibition of autophagy could be considered a strategy to ‘brown’ adipose tissues and enhance whole-body energy consumption to combat obesity. It is important to note that these findings should be interpreted with caution because many of the studies have been conducted on rodent models (especially mice) in which adipose tissue plasticity may be significantly higher than in humans and is based on species-specific neuroendocrinological mechanisms [60].

Liver tissue

The role of hepatic autophagy in the glucose homeostasis

Autophagy plays an important role in glucose homeostasis [93]. In this regard, hepatic autophagy is vital for the retention of energy balance, because one of the specific roles of the liver is to control homeostasis of glucose via retaining the concentration of blood glucose in the normal range and autophagy has an important part in this mechanism [94, 95]. During nutrient starvation, during short-term fasting, autophagy in the liver causes the production of glucose via the degradation of glycogen in the glycogenolysis pathway. But, in the prolonged fasting state, when the glycogen in the liver is depleted, hepatocytes produce glucose via the gluconeogenesis pathway [96,97,98]. Due to the role of autophagy in the liver in maintaining the blood glucose concentration, the results of a study on mice indicated that acute ATG7 deletion in the fasting state was associated with apoptosis induction in the liver tissue, reduction in the liver size, depletion of the stores of glycogen and lipid in the liver, and severe hypoglycemia, suggesting that autophagy is essential for the endurance of fasting, maintaining blood glucose, and survival [99]. In addition, Yang et al. revealed that in mice that were genetically or dietary obese, following the expression of ATG7 in the liver, insulin sensitivity was increased and glucose homeostasis was improved [100]. On the other hand, Liu et al. showed that in mice with hyperinsulinemia and insulin resistance induced by HFD, the expression of several important autophagy genes was inhibited. On the contrary, they showed that hepatic autophagy activity was enhanced in the group with insulin deficiency induced by streptozotocin (STZ), suggesting the suppression of autophagy in the state of hyperinsulinemia/insulin resistance [101].

Moreover, hepatic autophagy leads to the breakdown of proteins, and the resulting amino acids are converted to glucose in the gluconeogenesis pathway [102]. Generally, autophagy is inhibited by insulin and amino acids after food consumption, but in fasting states, it is stimulated through glucagon and the deficiency of amino acids [101]. Insulin inhibits hepatic autophagy via activating mTOR. The overabundance of nutrients is related to the enhanced activation of mTORC1 in the liver. Insulin can also downregulate numerous ATGs [103]. Unlike insulin, in fasting conditions, glucagon plays an important role in increasing blood glucose through the activation of glycogenolysis and gluconeogenesis and the inhibition of glycogenesis [104, 105]. In addition, glucagon activates autophagy by affecting the liver cells via increasing the number and size of autophagic vacuoles and upregulating lysosomal enzymes, acid phosphatase, and cathepsin D [106, 107]. It is suggested that glucagon via AMPK-mediated inhibition of mTORC1 activity, upgrades hepatic autophagy as well [108]. Nevertheless, the role of insulin is more important than glucagon in relation to controlling hepatic autophagy, because even at elevated plasma levels of glucagon, autophagy is induced only when blood insulin levels drop [109]. Taken together, hepatic autophagy is required to maintain the body's energy in a fasting state and insulin plays an important role in regulating the autophagy pathway.

Pathophysiological role of autophagy in hepatic steatosis and NAFLD

NAFLD, as the most prevalent chronic liver disease, is closely linked to hepatic insulin resistance and is observed in approximately 25% of the population. It includes a spectrum of hepatic disorders from a simple intrahepatic accumulation of TG or steatosis to fibrosis, cirrhosis, and hepatocellular carcinoma at advanced stages [110,111,112,113]. Insulin resistance and fat accumulation in the liver are among the most important factors involved in the pathogenesis of NAFLD [114]. Hepatic steatosis is the first stage in NAFLD pathogenesis, which is usually benign, and is an abnormal condition caused by the over-accumulation of LDs containing TG in hepatocytes. Lipophagy prevents the development of hepatic steatosis by degrading LDs [115,116,117,118]. This disorder is the result of an imbalance in the de novo synthesis of fatty acids in the hepatocytes or an increase in the number of fatty acids entering the liver, leading to an enhanced TG synthesis, or reduced secretion of very-low-density lipoprotein (VLDL) from the liver [119]. As a result of insulin resistance in adipose tissue, lipolysis is enhanced and leads to increased levels of free fatty acids (FFAs) in the liver. Due to the inhibition of autophagy by hyperinsulinemia and the role of autophagy in the breakdown of TGs, hepatic steatosis is worsened [120].

Studies have shown that in mice with HFD, the deletion of the Tfeb, ATG7, and ATG14 genes in hepatocytes, which encode vital autophagy regulators, as well as the deletion of the Atg5 gene in myeloid or endothelial cells, is associated with increased lipid accumulation in the liver and the development of NAFLD [121,122,123]. Overall, the inhibition of autophagy may be considered one of the causes of NAFLD. In this regard, during insulin resistance, the resulting hyperinsulinemia leads to the inhibition of autophagy via the activation of mTOR and the reduction of FOXO1 [124]. Research has indicated that dyslipidemia and steatosis are related to the impairment of FOXO, which is a transcription factor and the main intracellular target of insulin [124, 125]. FOXOs activate autophagy by inducing the expression of autophagy genes [126]. For example, the overexpression of FOXO1 in the Hepa1c1c7 cells prevents the inhibition of major autophagy genes by insulin [101]. Because FOXOs stimulate the degradation of LDs by activating lipophagy, a defect in their function may lead to the development of NAFLD [127].

One of the important regulators of the autophagy pathway is the transcription factor EB (TFEB) [128]. This transcription factor controls the expression of genes implicated in the regulation of lipid metabolism in the liver (such as oxidation of fatty acids and synthesis of ketone bodies). It also activates autophagy by inducing related genes [129, 130]. In this regard, metformin, which is a drug known to enhance insulin sensitivity, reduced insulin resistance and hepatic steatosis in a NAFLD mouse model induced by HFD by promoting autophagy in a TFEB-dependent manner [131, 132]. In obese animals, the absence of TFEB leads to disruption in the catabolism of lipids, whereas its high expression has the opposite effect [133]. Therefore, the suppression of TFEB and consequently the inhibition of autophagy lead to the accumulation of lipids in hepatocytes and induce steatosis [128]. Generally, the stimulation of hepatic autophagy by activators may have therapeutic potential and minimize liver impairment and NAFLD.

The role of autophagy in the regulation of hepatic lipid and lipoprotein metabolism and its importance in insulin resistance

The hepatic failure of lipid metabolism certainly contributes to the insulin resistance in this tissue and in the entire body tissues. Of all underlying mechanisms, autophagy impairments lead to the toxic accumulation of lipids, e.g., TG in the liver. In this regard, based on in vitro and in vivo studies, it is shown that the lack of some important autophagy genes such as ATG7 and ATG5 leads to the accumulation of TG levels in the liver; conversely, the overexpression of ATG7 leads to a decrease in the hepatic TG [100, 122]. In addition to ATG7 and ATG5, Xiong et al. revealed that ATG14, as an important factor in initiating autophagy, plays a critical function in the homeostasis of hepatic lipid metabolism. The overexpression of ATG14 in the liver of LTKO mice was shown to increase autophagy activity and decrease TG levels. Furthermore, the authors recognized one cluster and a single insulin response element (IRE) in the promoter of the ATG14 gene which regulates the promoter activity of this gene [122]. Additionally, Li et al. showed that the dysregulation of miR199a-5p, as an important regulator in hepatic glucose and lipid metabolism, inhibited the expression of ATG14 and suppressed autophagy, which can lead to impaired hepatic insulin sensitivity and ultimately insulin resistance [134]. Based on the fact that the combined effects of mTOR and insulin lead to the regulation of autophagy, Liu et al. reported that insulin resistance and hyperinsulinemia were induced in mice fed an HFD, which in turn inhibited hepatic autophagy [101]. The results of the study by Ghareghani et al. revealed that HFD in mice leads to enhanced expression of lipogenic genes and the impairment of the autophagy pathway. Moreover, in the HepG2 cell line, autophagy suppression via chloroquine increased lipogenesis, and the induction of autophagy by rapamycin, as a specific inhibitor of mTOR, decreased lipogenesis [135, 136]. The activation of autophagy by rapamycin improved insulin resistance and hepatic steatosis in animal models of T2DM, which was associated with a reduction in the levels of TG and fatty acids in the liver tissue [137]. However, Di Ma et al. indicated that liver autophagy deficiency by FIP122 knockout did not develop steatosis in both control and HFD-fed mice. Decreased lipid accumulation in the liver of these animals can be due the entire reduced expression of genes implicated in lipid metabolism including lipogenesis [138].

FFAs are other factors involved in the induction of hepatic insulin resistance via several mechanisms, including PKC-δ, phosphorylation of Akt, IRS-1, insulin receptor, and de novo lipogenesis [139, 140]. In the context of the autophagy response to FFAs, Tan, et al. conducted a study on cells treated with palmitic acid, a saturated fatty acid, showing that the autophagic flux level was increased, whereas oleic acid, a monounsaturated fatty acid, had no such effect [141]. On the other hand, Mei et al. reported that oleic acid induced autophagy while palmitic acid decreased it in primary mouse hepatocytes and HepG2 cells [142]. However, both studies demonstrated that autophagy inhibition by drug or ATG7 knockdown led to the obvious exacerbation of the toxic effects of palmitic acid and leads to further apoptosis in the palmitate-exposed cells, suggesting that autophagy acts as a protective mechanism against lipotoxicity. Fatty acid metabolites such as TG and DAG, involved in insulin resistance, trigger autophagy by which these are metabolized, raising the possibility that autophagy modulates liver insulin resistance by reducing TG stores [143].

In addition to TG, studies have documented that autophagy is implicated in lipogenesis, cholesterol efflux, ketogenesis, and oxidation of fatty acids [144]. For example, the accumulation of free cholesterol in hepatocytes leads to the dysfunction of lysosomes and the inhibition of hepatic autophagy; thus, the induction of cholesterol 7α-hydroxylase, as a rate-limiting enzyme in the biosynthesis of bile acids from cholesterol, leads to increased bile acid synthesis and decreased Akt/mTOR signaling, and ultimately activates autophagy [145,146,147].

Moreover, in relation to ketogenesis and autophagy, Saito et al. demonstrated that upon fasting, the loss of ATG5 or ATG7 in the liver disrupts the production of ketone bodies, indicating the importance of autophagy in the production of ketone bodies in fasting [144]. In addition, autophagy may control the level of VLDL particles, which are rich in TGs created in the liver [97]. The amount of VLDL and TG secretion from the liver depends on the availability of apolipoprotein B100 (apoB100), which is fundamentally produced via hepatocytes. Some studies have shown that the destruction of apoB100 is mediated by autophagy [148,149,150]. Andreo et al. showed that for the destruction of apoB100 in the insulin-stimulated hepatocytes, autophagy and class II PI3K are necessary. Additionally, the production of VLDL and apoB100 was excessive in those with insulin resistance [151]. Following the inhibition of autophagy by genetic and/or pharmacological factors, aberrant fat accumulation occurs in the liver, leading to insulin resistance [152]. The absence of lipophagy in the liver cells blocks the hydrolysis of TGs into FFAs and leads to steatosis and insulin resistance [53].

Autophagy and insulin resistance in liver tissue

Under the feeding conditions, insulin is released from pancreatic β-cells, and by acting on muscle and adipose tissues, it increases energy storage in these tissues in the form of glycogen and TGs, respectively [153]. Following the binding of insulin to its receptors in hepatocytes, the PI3K/Akt pathway is stimulated. Akt subsequently activates mTORC1 and suppresses FOXO (which is involved in the initiation of autophagy), leading to the inhibition of autophagy by insulin [34, 154]. One of the mechanisms by which insulin can block autophagy is the suppression of the vps34 gene, which is required for the initiation of autophagy, as well as ATG12 and GABA type A receptor-associated protein like 1 (gabarapl1) genes, which are necessary for the formation of complex I and II of autophagy, respectively. The expression of these genes in hepatocytes is FOXO1-dependent [101]. The results of study on mice with insulin resistance and hyperinsulinemia induced via HFD indicated that the hepatic autophagy and the expression of several main autophagy genes were suppressed [101]. Interestingly, autophagy was found to affect insulin signaling such that hepatic autophagy impairment promoted obesity and induced insulin resistance [46]. The results of an in vitro and in vivo study revealed that ATG7 suppression in the liver becomes an impairment in insulin sensitivity, and the recovery of ATG7 gene expression in obese mice leads to increased hepatic insulin activity [100, 118]. Figure 3 shows the insulin signaling in liver tissue under physiological and pathological conditions (3.2.a and 3.2.b). With regard the intercalated relation between insulin signaling and autophagy, hence, the interaction should be further studied in the setting of insulin resistance.

Autophagy and ER stress in liver tissue and their relationship with insulin resistance

Studies have revealed that following intense ER stress, the IRE1–JNK pathway is activated, which subsequently suppresses insulin receptor signaling in the liver cells [94, 155]. The secretion of fatty acids from adipocytes in obesity induces the unfolded protein response (UPR) pathway and by activating the JNK and inhibiting the expression of chaperone proteins and ER-associated degradation (ERAD), insulin resistance occurs in the liver [94]. Furthermore, some findings propose that the upregulation of tribbles-related protein 3 (TRB3), a pseudokinase that suppresses the activation of Akt, via the PERK pathway in diabesity could contribute to the induction of hepatic insulin resistance by ER stress [156, 157]. In addition to the ERAD, which degrades misfolded and unfolded proteins, autophagy is also induced by ER stress and UPR [158]. Autophagy induced by ER stress is considered a mechanism for the degradation of misfolded proteins that cannot be decomposed by ERAD [100]. ER stress induces autophagy via several mechanisms such as the IRE1–JNK pathway, ER Ca2+ leakage, the assembly of pre-autophagosome, the enhancing of AGT1 kinase activity, and the upregulation of ATG genes [159,160,161,162,163]. The results of a study on the rat model of NAFLD treated with an inhibitor of JNK showed that the expression levels of ATG3 and ATG5 were decreased, insulin resistance was reduced, and NAFLD was improved [164]. Nevertheless, it has been shown that the reduction of hepatic autophagy activity causes ER stress and insulin resistance [165]. In this regard, Yang et al. reported that in a starvation state, the subsequent imperfect hepatic autophagy in obesity and the suppression of AGT7 induce ER stress and insulin resistance, leading to the assembly of lipids in the liver [100]. Furthermore, it was reported that in the livers of murine models and patients with NAFLD, the autophagic pathway is impaired, which could be due to the fact that increased ER stress causes apoptosis [166]. Overall, the increase in ER stress and the decrease in autophagy activity may lead to impaired insulin signaling and insulin resistance in liver tissue.

Autophagy, mitochondrial dysfunction, inflammation, and oxidative stress in the liver tissue and their links with insulin resistance

Mitochondrial dysfunction is a hallmark of chronic and acute liver disorders, such as NAFLD, alcoholic liver disease, and viral hepatitis [167, 168]. Mitochondrial dysfunction is also involved in the development of diabetes and insulin resistance, as mitochondria produce a high amount of ROS and oxidative stress plays an important role in insulin resistance [169,170,171]. One of the pathways by which ROS causes insulin resistance is JNK activation [172]. It was proven that FFAs promote mitochondrial dysfunction and accumulate in the liver, thus inducing hepatic insulin resistance [173]. Upon the accumulation of ROS and the elevation of oxidative stress as a result of increased proinflammatory factors and FFAs, the signaling pathways of various stress-sensitive serine/threonine kinases such as IKKβ, are activated and several targets of these kinases including IRS-1 and IRS-2 and the insulin receptor are phosphorylated at serine and threonine sites, resulting in decreased insulin function and insulin resistance [14, 174].

Autophagy is essential in limiting ROS generation by removing damaged mitochondria. On the other hand, autophagy is induced by ROS at several levels: In the initiation stage, through the modulation of AMPK and mTORC1 activity, in the nucleation phase, through the activation of protein kinase D and caveolin-1, and in the elongation step, through inducing the activation of ATG4 [175]. Accordingly, the suppression of autophagy leads to increased production of ROS [176]. Furthermore, the inhibition of autophagy triggers the production of inflammatory cytokines IL-1β and TNF-α, by blocking NF-κB and inflammasome [177]. Studies have revealed that autophagy can negatively regulate the activation of the inflammasome via several mechanisms, including the elimination of damaged mitochondria, which resulted in a drop in ROS levels and the consequent inhibition of the inflammasome, p62-dependent breakdown of the inflammasome, and finally, the sequestration of pro-IL-1β [178]. It was reported that IL-1β disrupts insulin signaling and leads to insulin resistance in hepatocytes via decreasing IRS-1 expression [179]. In this regard, it was demonstrated that the alleviation of inflammation by removing Kupffer cells form the liver restored autophagy and improved insulin resistance in HFD-fed mice [180, 181]. Therefore, the reduction of liver macrophages can be a suitable strategy to increase hepatic autophagy and reduce liver inflammation, insulin resistance, and related metabolic diseases such as NAFLD and T2DM.

Skeletal muscle tissue

Muscles are responsible for the body's movement and protection. However, continuous muscle contractions cause ROS production and damage muscle cell proteins. Thus, these cells need specific mechanisms to remove damaged proteins and organelles and supply amino acids from proteins [182]. Along with mechanisms such as proteasomal degradation, autophagy is one of the processes that can help to eliminate damaged proteins, prevent their accumulation, and convert them into amino acids.

Insulin, as the most important regulator of blood glucose, stimulates glucose uptake mainly via skeletal muscle. Muscle insulin resistance is considered an essential component of whole-body insulin resistance since skeletal muscle is responsible for about 60–70% of insulin-stimulated glucose disposal in the entire body [183]. Insulin-stimulated glucose absorption in skeletal muscle is significantly reduced under insulin-resistant conditions, allowing for the development of T2DM. However, autophagy can improve insulin resistance in skeletal muscle [184].

Autophagy, insulin signaling, and insulin resistance in skeletal muscle tissue

Crosstalk between autophagy and insulin signaling is of particular significance. It was demonstrated that the inhibition of autophagy by chloroquine, bafilomycin, or the inactive ATG5 mutant diminished glucose uptake in the L6 skeletal muscle stimulated by insulin. In addition, considerable reductions were observed in the phosphorylation levels of Akt and IRS-1 [86]. Numerous studies have described the importance of autophagy in the insulin-resistant skeletal muscle in vivo. In HFD-fed mice and animal models of polycystic ovary syndrome (PCOS) with hyperinsulinemia and insulin resistance, autophagy reduced in skeletal muscle [86, 185]. However, six weeks of HFD feeding resulted in enhanced autophagy markers (LC3II and beclin), indicating that autophagy increased as a compensatory response to early insulin resistance [86]. Interestingly, genetic animal models with continuous hyperactive autophagy manifest more insulin sensitivity on the HFD challenge, as evidenced by increased phosphorylated Akt in the gastrocnemius skeletal muscle [93].

It is noted that insulin sensitizers, such as adiponectin, promote the autophagy pathway in skeletal muscle [86]. Exercise, which is known to improve insulin resistance, partly functions through autophagy [186]. In addition, Song et al. showed that dehydroepiandrosterone activates mTORC1 and inhibits autophagy, leading to insulin resistance in the skeletal muscle of mice with PCOS [185]. Shi et al. reported that dihydromyricetin, a flavonoid component of herbal medications, improves the skeletal muscle insulin sensitivity by inducing autophagy via the AMPK–PGC-1α–sirtuin 3 (Sirt3) signaling pathway [187]. The specific knockout of ATG7 in skeletal muscle unexpectedly improved glucose uptake and insulin resistance following HFD feeding [54].

The skeletal muscle of T2DM patients with hyperinsulinemia exhibited a reduction in autophagy, which was demonstrated by the reduced expression of SQSTM1/p62, ATG14, GABARAPL1, RB1 inducible coiled-coil 1 (RB1CC1)/ family-interacting protein of 200 kDa (FIP200), and WD repeat domain phosphoinositide-interacting protein 1 (WIPI1)genes, decreased levels of SQSTM1/p62, LC3BII, and ATG5 proteins, as well as the elevated phosphorylation of FOXO3a [188]. These data indicate that insulin resistance is associated with autophagy inhibition in muscles while increased insulin sensitivity had inverse effects [189]. Conversely, Kruse et al. did not find any changes in the mRNA and protein levels of the autophagic markers of the skeletal muscle in T2DM patients, suggesting that markers of autophagy are probably adapted to hyperglycemia in skeletal muscle in T2DM patients [189]. Regarding the in vivo impact of insulin on autophagy, it is noted that the glycemic status, as a crucial factor, as along with insulin affect autophagy in patients with diabetes [190]. Insulin infusion led to a decreased LC3BII/I ratio in obese and lean individuals, but not in diabetic patients, indicating that insulin is capable of inhibiting autophagy in conditions other than diabetes [189]. In this context, the results of the study by Lv et al. are noteworthy. They investigated autophagy alterations in hyperglycemic conditions with or without hyperinsulinemia, by using both animal models of hyperglycemia, i.e., hyperglycemia by glucose infusion and hyperglycemia induced by STZ. In STZ-treated rats, which present hyperglycemia and low insulin, autophagy proteins (LC3, ATG5, ATG12, and BECN1) were significantly elevated in the gastrocnemius muscle, which contributed to the increase of p-FOXO3a and the subsequent repression of mTOR. In contrast, in animals with glucose infusion hyperglycemia, which present hyperinsulinemia, autophagy markers, including LC3-II/I, ATG7, and BECN1 decreased in gastrocnemius muscle, indicating that insulin in the lack of insulin resistance can suppress autophagy in skeletal muscle [191]. It appears that the status of insulin and glycemia, as well as insulin sensitivity, altogether create a unified response that determines the stimulation or inhibition of the autophagy process.

Furthermore, the glucagon level, which is elevated in the presence of high insulin and insulin resistance state, is another main factor that regulates autophagy [192]. The effects of glucagon on the autophagy pathway are the opposite of insulin’s effects, i.e., glucagon activates autophagy. It has been found that chronic exposure to insulin diminishes the stimulating effects of glucagon on autophagy in hepatocytes [101], indicating that insulin is more important than glucagon in autophagy regulation. However, more research is required to clarify the in vivo effects of different variables on skeletal muscle autophagy. Overall, the autophagy function is balanced under normal conditions, whereas it is inhibited in the presence of insulin. Nevertheless, it may be increased as a compensatory response to maintain homeostasis. Figure 3 shows insulin signaling in modules under physiological and pathological conditions (3.3.a and 3.3.b).

Autophagy and metabolism of glucose and lipids in skeletal muscle tissue

Insulin resistance is associated with elevated levels of glucose, circulating fatty acids, and TG, as well as lipid accumulation, which is linked to metabolism abnormalities in the skeletal muscle [193]. It is well established that an HFD results in the accumulation of lipid derivatives, such as triacylglycerol, diacylglycerol, and ceramide, in skeletal muscle, leading to insulin resistance [194]. Several studies support the role of autophagy in lipid metabolism, suggesting that it is impaired in pathological conditions such as insulin resistance [86, 185, 195]. It was demonstrated that p62, an autophagy receptor targeting cargo to autophagosomes, is found on LDs, which indicates that lipophagy participates in the removal of TG from muscle cells such that the treatment of fatty acid-exposed muscle cells with rapamycin (an autophagy activator) reduced lipid accumulation while bafilomycin (an autophagy inhibitor) showed the opposite effect [196]. On the other hand, lipids can modulate the autophagy process in skeletal muscle cells. It was demonstrated that HFD globally reduces the expression of autophagy genes in the soleus (SOL) muscle [197]. Palmitate, a saturated fatty acid with high concentrations in circulation, and known to promote insulin resistance in skeletal muscle, impaired the autophagic flux in C2C12 cells [195]. These data suggest that autophagy impairment contribute to lipid accumulation and thereby insulin resistance in muscles. In contrast, Morales-Scholz concluded that autophagy is neither implicated in lipid accumulation nor in the development of insulin resistance in skeletal muscle. In this study, feeding rats with a high saturated-fat diet (HSFD) for 16 weeks did not change the autophagic markers in their skeletal muscle, but autophagy defects were seen in the liver [198]. The difference between oxidative muscles and glycolytic muscles in autophagy is of particular significance. While autophagic proteins (LC3-I, LC3-II, and SQSTM1) were higher in the oxidative muscle compared to the glycolytic muscle, autophagic flux was low in the oxidative muscle [199]. In female mice fed an HFD, p62 and ATG12-5 accumulation was induced in the oxidative muscle (SOL) but not in the glycolytic muscle (plantaris) [200]. It is noted that HFD induces the accumulation of diacylglycerols and ceramides in oxidative muscles SOL and extensor digitorum longus (EDL) but not in the glycolytic muscle epitrochlearis (Epit) [201]. It appears that the importance of autophagy with regard to lipid accumulation in muscles depends on the muscle type.

In addition, it has been shown that autophagy activity is related to glucose homeostasis. It was found that the glycogen stores of the skeletal muscle were mobilized through the autophagy degradation pathway in newborn rats [109]. The hexokinase (HK) enzyme, which catalyzes the first reaction of glycolysis, exerts a modulatory effect on autophagy [202]. Roberts et al. demonstrated that HKII suppresses the autophagy inhibitor mTORC1, thereby activating autophagy [203]. In C2C12 cells under the condition of low glucose concentrations, HKII was upregulated, which is associated with the induction of autophagy [204]. It appears that HKII, as a regulatory molecule, plays a crucial role in the metabolic switch from glycolysis to autophagy during glucose starvation. Given that HKII is regulated by insulin, its possible downregulation during insulin resistance leads to impairments in the autophagy pathway.

It should be noted that skeletal muscle autophagy may be altered in response to pathological conditions manifesting metabolic syndrome. For example, in NAFLD, myocellular autophagy is enhanced as a metabolic response to hyperammonemia [205], or in animals with hypertension, autophagy markers, including LC3I, ATG7, and lysosome-associated membrane protein 2 (LAMP2), are increased in skeletal muscle [206]. It can be concluded that the metabolism of muscle cells is influenced by insulin resistance, which is associated with autophagy dysregulation [205, 206].

The role of myokines in autophagy-related insulin resistance

Myokines are peptides or cytokines that are synthesized and secreted by skeletal muscle, with autocrine, paracrine, and endocrine activities. They include myonectin, myostatin, apelin, fibroblast growth factor 21 (FGF21), brain-derived neurotrophic factor (BDNF), irisin, IL-15, IL-6, and other similar factors [207]. Myokines are potentially involved in regulating metabolism, autophagy, and insulin resistance as well as crosstalk between skeletal muscle and other tissues [208].

Myostatin is known as a negative regulator of skeletal muscle mass. In addition, it plays a role in the pathogenesis of insulin resistance. Its circulating levels are increased in obese patients with insulin resistance, and it has a negative correlation with adiponectin levels and insulin sensitivity indices [209]. In C2C12 skeletal muscle cells, it was found that myostatin exerted deleterious effects on the factors involved in the insulin signaling pathway, including IRS-1, glycogen synthase kinase-3 (GSK3), Akt, GLUT4, AMPK, and PGC1α [210]. Furthermore, it was reported that myostatin potentiates the autophagy process in C2C12 cells, accounting for muscle mass loss and muscle atrophy in these cells [211]. Studies have shown that myostatin inhibition by gene silencing [212], anti-myostatin oligomers [213], or by anti-myostatin antibodies [214] improves insulin resistance. It appears that the improvement of insulin sensitivity by myostatin suppression is due to the increased muscle mass and insulin signaling reinforcement rather than autophagy regulation.

Irisin is primarily produced by the skeletal muscle. Both autophagy and irisin increase in skeletal muscle following exercise and it seems that both mediate the beneficial effects of exercise on skeletal muscle insulin resistance [215]. In insulin-resistant C2C12 skeletal muscle cells, it was found that irisin could induce the autophagy process through the p38 MAPK/PGC-1α axis, improving insulin signaling and glucose uptake [216].

Several studies have reported that CTRP15 is related to insulin resistance in patients with metabolic syndrome [217], coronary artery disease [218], T2DM [219], and PCOS [220]. Its serum levels significantly increased in individuals during exercise [221]. It has been demonstrated that CTRP15 negatively regulates liver autophagy via Akt/mTOR signaling, as evidenced by reduced Atg expression and p62 degradation [222].

IL-15 and IL-6 are cytokines secreted by skeletal muscle cells, addition to the immune cells during immune responses, and are involved in the pathogenesis of autoimmune diseases and insulin resistance [223]. IL-15 has been shown to regulate skeletal muscle mass by suppressing the ubiquitin proteolytic process. In addition, it directly stimulates glucose uptake in muscle cells [224, 225]. High levels of IL-15 contribute to the improvement of insulin resistance in postmenopausal women with metabolic syndrome [226]. IL-15 triggers autophagy in the immune cells, mediating homeostasis and the survival of these cells [227, 228]. IL-6 is a pleiotropic cytokine with respect to insulin resistance; while numerous studies have indicated its association with insulin resistance [226,227,228,229], others have reported its insulin-sensitizing effects [230, 231]. It has been found that IL-6 inhibits starvation-stimulated autophagy in pro-monocytic U937 cells [184]. Nevertheless, it is still unclear whether IL-15 and IL-6 trigger autophagy in order to regulate insulin signaling in skeletal muscle cells.

Overall, myokines provide crosstalk between skeletal muscle and other tissues and serve functions in all tissues. The available data are not sufficient to fully describe their contribution to autophagy-related insulin resistance, and since skeletal muscle mass is crucial in insulin sensitivity and a high autophagy rate leads to muscle loss, it should be considered to gain a correct understanding of the role of myokines in autophagy-related insulin resistance.

Autophagy, mitochondrial dysfunction, ER stress, and oxidative stress in skeletal muscle tissue and their links with insulin resistance

A growing body of evidence links mitochondrial dysfunction and oxidative stress to insulin resistance [229]. Particularly, in the muscle, it has been shown that elevated ROS levels stimulate the p38 mitogen-activated protein kinase (MAPK)/JNK/ERK pathway, which is involved in insulin resistance. Furthermore, increased levels of ROS activate mitophagy in the skeletal muscle such that the treatment of myotubes with H2O2 augments the levels of phosphorylated ULK1 and LC3 proteins [230]. Autophagy can mutually modulate mitochondria function and oxidative stress. In the skeletal muscle of ATG7-deficient mice, the protein carbonyl level, as a biomarker of oxidative stress, was increased and the ATP content and cytochrome oxidase activity were reduced, suggesting that autophagy deficiency leads to oxidative stress and mitochondrial impairments in the muscle, which is associated with reduced insulin resistance [54]. ATG7 deficiency in the muscle is associated with the induction of FGF21. FGF21 is a mitokine that increases browning WAT and fatty acid oxidation, thereby improving insulin sensitivity [54]. Kim et al. reported that mitochondrial dysfunction in the muscle with ATG7 deficiency improves insulin resistance. This finding is in contrast with the results of previous studies, which stated that mitochondrial dysfunction and autophagy impairment exacerbate insulin resistance [47, 184, 185]. It appears that the outcomes of autophagy deficiency depend on (1) where it occurs and (2) the component of the autophagy mechanism that is disrupted.

Considering the importance of muscle mass in whole-body responses to insulin, individuals who are prone to muscle mass loss are more likely to develop insulin resistance [231]. Oxidative stress in muscles initiates the process of muscle atrophy via activating autophagy, thereby decreased insulin resistance [232]. For example, neuronal nitric oxide synthase (nNOS) reduces muscle mass by activating oxidative stress and increases the expression of atrophy-related ubiquitin ligase genes (atrogin 1 and MuRF1) [233]. Moreover, MAPK induces autophagy during oxidative stress [234]. The formation of autophagosomes in myofibers is associated with atrophy and muscular dystrophy [235]. In addition, it has been indicated that the overexpression of PGC-1α (a mitochondrial biogenesis inducer) in skeletal muscle improves age-associated insulin resistance and reduces age-induced autophagy, thereby preventing muscle mass loss during aging [238]. However, the muscle-specific knockouts of autophagy genes, including Atg5−/− and Atg7−/−, lead to muscle atrophy, presenting a myopathy-like phenotype [236, 237]. All the data demonstrated that proper muscle function requires the precise regulation of autophagy.

ER stress was observed in skeletal muscle with insulin resistance. It has been shown that ER stress disrupts insulin signaling in skeletal muscle and therefore reduces glucose uptake, which is mediated by tribbles-like protein 3 (TRBP3) [156]. ER stress is also known as an autophagic inducer, since the three main arms of ER stress, i.e., PERK, IRE1α, and ATF6α, trigger autophagy by activating JNK, X-box binding protein 1 (XBP1), and ATF4 [238]. It has been found that the treatment of C2C12 skeletal muscle cells with an ER stress inducer augments the conversion of LC3-I to LC3-II and decreases p62. In these cells, PKC-ϴ is demonstrated to mediate the ER stress-induced effects on autophagy [239]. The activation and membrane localization of PKC-ϴ might contribute to identifying membrane vesicles that form autophagosomes. Another candidate molecule that links ER stress to autophagy is skeletal muscle- and kidney-enriched inositol polyphosphate 5-phosphatase (SKIP). This phosphatase, which is upregulated under ER stress, negatively regulates insulin signaling by dephosphorylating PI3, and therefore, stimulating autophagy [240]. In general, ER stress is followed by autophagy activation, as a protective mechanism to remove misfolded proteins and thus relieve ER stress [238], providing cell survival. Additionally, the alleviation of ER stress is one of the main mechanisms by which autophagy lowers insulin resistance [93]. It was found that the genetic hyperactivation of autophagy improved HFD-induced insulin resistance in insulin-responsive tissues such as skeletal muscle, which was attributed to relieved ER stress, as evidenced by reduced levels of ER stress markers CHOP and ATF-6 factor in the skeletal muscle, liver, and adipose tissue [93]. In support of these findings, it was shown that ER stress inducers reversed the beneficial effects of autophagy hyperactivation in the aforementioned tissues [93].

However, autophagy acts as a defense response against ER stress, and depending on the degree of insulin resistance, this process is affected in different ways [61]. It should be noted that autophagy dysregulation may contribute to ER stress-mediated insulin resistance. In this regard, it is demonstrated that autophagy inducers (rapamycin or adiponectin) relieved ER stress and insulin resistance in skeletal muscle L6 cells while autophagy suppression had the opposite effect [241]. Therefore, it is concluded that under insulin resistance conditions, dysfunction of autophagy makes cells more vulnerable to ER stress.

Skeletal muscle insulin resistance is linked to inflammation, and the increase of inflammation enhances skeletal muscle insulin resistance [242]. IL-1, TNF-α, C-reactive protein (CRP), and monocyte chemoattractant protein-1 (MCP-1) are associated with skeletal muscle insulin resistance [243]. Insulin signaling is mainly disrupted by inflammation, ER stress, oxidative stress, the activation of the JNK and NF-кB pathways, and the increase of skeletal muscle insulin resistance. The JNK pathway and TNF-α disrupt insulin signaling by phosphorylating IRS-1 on the serine residue [244, 245]. IL-1 is demonstrated to activate the IKK/NF-кB pathway in the skeletal muscle, decrease IRS-1 activity, and enhance skeletal muscle insulin resistance. Moreover, IL-6 increases the expression of the suppressors of cytokine signaling (SOCS) 1 and SOCS3, leading to the degradation of IRS-1 and the development of insulin resistance [246]. Inflammation can also increase nitric oxide production, inhibit the PI3K-Akt pathway, and promote insulin resistance [244]. Overall, it seems that autophagy, mitochondrial dysfunction, ER stress, inflammation, and oxidative stress in the skeletal muscle are closely related to each other as a network and can all be affected by insulin resistance.

Autophagy abnormality in β-cells under metabolic disorders

Metabolic disorders, such as obesity and diabetes, affect β-cell function through various mechanisms, including the dysregulation of autophagy. In these conditions, excess nutrients compel β-cells to secrete more insulin to maintain normal glucose levels. This increased demand on β-cells overloads the ER, leading to ER stress, poly-ubiquitinated protein aggregates, incomplete insulin processing, and/or irregularities in insulin secretion [247]. It has been established that blocking autophagy through chemicals or short hairpin RNAs (shRNAs) enhances the death of β-cells under ER stress, while treatment with rapamycin, an autophagy inducer, increases the viability of β-cells in the presence of ER stress, which demonstrates the protective role of autophagy against ER stress in β-cells [248]. Furthermore, evidence indicates that an excess of lipids and the resulting lipotoxicity, which is accompanied by metabolic abnormalities, significantly contribute to the impairment of β-cells. The expression of genes involved in lipid metabolism, oxidative stress, and apoptosis exhibits an abnormal pattern within the β-cells of individuals with type 2 diabetes [249]. In these cells, the accumulation of lipid droplets is linked to the downregulation of transcription factor EB (TFEB), which is a crucial modulator of autophagy. This, in turn, led to a decrease in the levels of the lysosomal biomarker LAMP2 [249]. Hyperglycemia is another primary factor that affects β-cells in metabolic disorders. It seems that chronic hyperglycemia or glucotoxicity affects lysosomal degradation in islets [250]. Lipid overload and high glucose levels reduce the expression levels of TFEB, LAMP1, and LC3, as well as p62 aggregation in INS‐1 cells, indicating that autophagy is dysregulated by hyperlipidemia and hyperglycemia [249]. Glucotoxicity also leads to increased production of ROS and mitochondrial abnormalities in β-cells [251]. It has been demonstrated that ROS, including H2O2, can induce autophagy as a protective mechanism [252]. In this context, when β-cells are exposed to amyloid polypeptides, ROS triggers an autophagy response to mitigate cellular injury. Conversely, the inhibition of autophagy worsens oxidative stress and β-cell damage [253]. Human islet amyloid polypeptide (hIAPP) is another key element involved in the pathogenesis of β-cell dysfunction in patients with type 2 diabetes [254]. As autophagy is responsible for clearing hIAPP, a deficiency in autophagy results in the accumulation of hIAPP, subsequently contributing to the development of diabetes [255]. It was found that the inhibition of autophagy exacerbates oxidative stress and β-cell damage induced by amyloid polypeptides [253]. Overall, reduced autophagy function resulting from genetic predisposition, aging, and obesity may contribute to β-cell impairment and diabetes. Therefore, the modulation of pancreatic β-cell autophagy is a promising approach for addressing diabetes related to hyperlipidemia, hyperglycemia, and the accumulation of islet amyloids.

Complications

Obesity and inflammation

Sarcopenic obesity has become a notable global health concern. The loss of muscle mass and fat accumulation in obesity causes an imbalance in myokines and adipokines, resulting in the development of insulin resistance and chronic inflammation [256]. Obesity, which is known as a condition of insulin resistance, is associated with chronic inflammation. It has been demonstrated that inflammatory factors, including IL-1, TNF-α, CRP, and MCP-1, are linked to tissue insulin resistance, and this relationship is mediated by the JNK and NF-кB pathways [256]. It was found that IL-1 and TNF-α disrupt insulin signaling by phosphorylating IRS-1 on the serine residue [257, 258]. Furthermore, in the progression of obesity, the imbalance between food intake and energy expenditure leads to autophagy suppression owing to the activation of mTOR signaling [77]. It seems that inflammatory factors are implicated in the autophagy dysfunction observed in insulin resistance-associated obesity. Consistent with this finding, it has been indicated that inflammatory agents, such as lipopolysaccharides, induce autophagy dysregulation, as evidenced by the decreased levels of ATG5, ATG6, and beclin 1, as well as increased p62 levels in macrophages [63, 259]. In addition, autophagy was impaired in the macrophages of HFD-induced obese animal models presenting chronic inflammation and insulin resistance [63]. In contrast, in the adipose tissue of obese individuals with moderate degrees of inflammation, an upregulation of autophagy markers was observed [257], which might be part of the defense response. Autophagy and inflammatory events are shown to be mutually linked. It is well established that autophagy influences IL-1β production via the inflammasome complex in neutrophils [258] and macrophages [259]. IL-1β is upregulated in macrophages that are Atg16L1-deficient [259]. Additionally, rapamycin (an autophagy activator) attenuates inflammatory cytokines, thereby improving insulin resistance [137]. Taken together, these findings demonstrate the interactions of autophagy and inflammatory agents and highlight the importance of autophagy in inflammation-regulated insulin resistance.

Nephropathy

Nephropathy is characterized by a progressive rise in albuminuria, blood pressure, hyperfiltration, and podocyte loss as well as a decline in the glomerular filtration rate [260]. Nephropathy represents the disruption of the glomerular filtration barrier (GFB) [261, 262].

Autophagy maintains glomerulus and tubule homeostasis by recycling macromolecules and damaged cytoplasmic components and serves as a renoprotective mechanism for renal cells against stressful conditions [263, 264]. Recent evidence suggests that an imbalanced autophagic process is implicated in glomerular dysfunction and tubulointerstitial pathologies in the kidneys under insulin resistance conditions [265, 266]. Podocytes have a high level of autophagy activity even under normal conditions, implying that they require a basal level of autophagy function to maintain the integrity, normal structure, and function of the kidney [260]. Recent research has revealed that alterations in autophagy contribute to podocyte detachment with consequent albuminuria and a decline in filtration capacity. For example, the podocyte-specific deletion of Atg genes leads to podocyte loss and accelerates glomerulopathy [267]. Numerous studies have reported that the autophagy response in podocytes is lower in insulin resistance conditions [266, 268]. High glucose concentrations in cultured podocytes lead to molecular changes including the prevention of the normal autophagy response by restricting the expression level of the beclin-1, LC3, and Atg12-5 complex. This dysregulation of basal autophagy diminishes the filtration barrier function of podocytes [260]. Several investigations have indicated that the autophagy response is declined in the podocytes of insulin-resistant patients and animal models. In particular, autophagy insufficiency was observed in the kidney biopsy of patients with diabetic nephropathy and the animal models of insulin resistance with renal accumulation of p62/SQSTM1 and decreased levels of lysosome-associated membrane protein type 2A (LAMP-2A), which was accompanied by dysregulated autophagy function, enhanced lysosome dysfunction, and apoptosis [267].

Retinopathy

The diagnosis of retinopathy is made on the basis of impaired retinal neuronal function, apoptosis of neuronal cells, especially amacrine cells, retinal ganglion cells, and photoreceptors, the thinning of the nerve fiber layer, pericyte loss, the breakdown of the blood–retinal barrier (BRB), the activation of Müller cells or reactive gliosis, and the overexpression of vascular endothelial growth factor (VEGF) and proinflammatory cytokines [269, 270]. Retinopathy is associated with decreased contrast sensitivity, slow adaptive response to dark, constricted visual fields, and finally, visual dysfunction [271].

There is increasing evidence that high basal autophagy is particularly maintained in the retinal pigment epithelium (RPE) and photoreceptor cells with normal retinal morphology and visual performance [269]. This baseline autophagy response reduces RPE and photoreceptor death as well as neural degeneration through the increased recycling of toxic rhodopsin. An inadequate autophagy flux leads to light-induced retinal degeneration. It has been reported that retinal autophagy is upregulated in individuals and mice with insulin resistance [270]. Autophagy enables retinal cells to counteract intracellular stress; however, in insulin resistance conditions, the autophagy machinery is a double-edged sword. Autophagic response in retinal cells may contribute to cell survival or the disruption of cellular homeostasis and cell death, depending on the level of stress and rBRB leakage. Autophagy activation in insulin resistance is a protective response against cell death in moderate stress, but it is an adverse response in excessive stress and contributes to cell death [272].

Müller cells or retinal glial cells (RGCs) cover all the retinal layers. The preliminary induction of autophagy prevented RGC death, but hyperglycemia-induced uncontrolled excessive autophagic response increased p62/SQTSM1 levels and led to an accelerated apoptotic rate and substantial VEGF formation [272]. The inhibition of the autophagy machinery by 3-methyladenine (3-MA) and chloroquine aggravated RGC apoptosis in a hyperglycemia environment while treatment with rapamycin improved the autophagic cargo degradation, and subsequently, promoted RGC viability [273].

Treatment of RPE cells with a high concentration of glucose accelerated ROS production, increased cell toxicity, reduced cell viability, and enhanced autophagic processes. These adverse impacts were eliminated by ROS scavengers and exacerbated by autophagy inhibitors in a dose- and time-dependent manner, indicating the protective role of autophagy in these cells [274]. In early retinopathy, autophagy activation attenuates pericyte loss as well as BRB disruption and enhances cell survival [269]. Pericytes are highly specialized cells that cover the retinal capillaries. As retinopathy develops and due to BRB leakage, autophagic death occurs. Pericyte loss leads to the obstruction of capillaries and microaneurysms [269].

Cardiomyopathy and vascular dysfunction

Cardiomyopathy is characterized by impaired cardiomyocyte contractility and performance [275]. There is evidence indicating that autophagy dysregulation is involved in cardiomyopathy; for example, cardiac ablation of the ATG5 gene leads to an increased incidence of cardiac dysfunction [276]. However, the cardiac autophagy activity in insulin resistance conditions has not been clearly described, and contradictory roles have been reported for cardiac autophagy in type 1 and type 2 diabetic animals [277]. Autophagic flux is enhanced in the heart of STZ-treated mice, as evidenced by the abundance of autophagic vacuoles or autophagosomes, the LC3-II/I ratio, and SQSTM1/p62 [277]. ATP deficiency or the accumulation of AMP in type 1 diabetic heart promotes AMPK activity in order to initiate autophagy, which results in cardiac failure [277]. Furthermore, treatment with chloroquine, an autophagy inhibitor, worsened cardiac diastolic and systolic performances in STZ-induced diabetic mice [277]. However, in db/db mice, as a model of T2DM, autophagy is disrupted in the step of autophagosome fusion with lysosome in heart tissue [277]. Moreover, the cardiac autophagy process was inhibited in obesity and HFD-induced insulin resistance, which was associated with increased levels of p62 protein and decreased levels of LC3II [278]. Several studies have revealed that the hyperactivation of autophagy in insulin resistance leads to cardiac cell degeneration and cytotoxicity [279]. Autophagy not only plays a crucial role in aberrant cardiac function but also contributes to vascular dysfunction and atherosclerosis pathogenesis. It has been shown that aortic dissection is exacerbated by ATG5 deficiency in vascular smooth muscle cells (VSMCs), which also prevents autophagosome formation and increases cell death [280]. Neointima development, atherogenesis, and accelerated VSMC senescence are all linked to defective autophagy in VSMCs [280]. It has been reported that chaperon-mediated autophagy is stimulated during initial pathogenic events, but it decreases upon atherosclerosis progression [281]. These data imply that basal autophagy functions as a protective mechanism to preserve the homeostasis of VSMCs and endothelial cells and prevents plaque formation, while excessive autophagy may exert the opposite effects.

Infertility

Autophagy plays a critical role in female reproductive function by regulating folliculogenesis, the survival of germ cells, implantation, the maintenance of placental physiology, pregnancy, and anovulation [282]. A larger number of autophagosomes were detected shortly after fertilization, indicating the necessity of autophagy after fertilization, which removes unnecessary cellular proteins and supplies the acquired amino acids for embryonic progression [283]. It has been reported that the expression of autophagy-related genes is downregulated in the endometrium of women with PCOS and insulin resistance, and metformin (an insulin-sensitizing drug) counteracts these alterations [284]. Autophagy dysregulation was also reported in primary human granulosa cells with insulin resistance [285]. On the contrary, autophagy was upregulated in the ovarian tissue of PCOS rats and humans [135]. It has been reported that the overactivation of autophagy contributes to the impairment of HFD-related spermatogenesis since treatment with chloroquine, an autophagy inhibitor, ameliorates spermatogenesis defects in animals [286]. Moreover, excessive autophagy was observed in semen samples obtained from obese individuals [286]. Overall, although autophagy function is not necessary for the reproductive system to work correctly, it may play a different role in pathological conditions, implying the dual role of autophagy.

Targeting autophagy in the treatment of diabetes

Table 1 indicates the Food and Drug Administration (FDA)-approved medications that possess a recognized indication on metabolic processes and have the potential to impact the autophagic pathway. Given that autophagy activity is inhibited in diabetic patients, new treatment strategies concentrating on autophagy restoration have been developed. To treat diabetic kidney disease (DKD), antioxidants can be used to combat oxidative stress, or chemical chaperons can be employed to lower ER stress and restore autophagy activity [287]. Reduced autophagy may be implicated in the development of DN when AMPK activation is reduced. Therefore, AMPK activation could be a target for reactivating autophagy. Reduced AMPK activity, and consequently, reduced cardiac autophagy are key factors in the progression of diabetic cardiomyopathy [84, 288]. In this regard, certain drugs, such as metformin and 5-aminoimidazole-4-carboxamide ribonucleotide, may alleviate DN by restoring autophagy. Furthermore, Zhou et al. showed that tea polyphenols improved rat heart function in diabetic cardiomyopathy and might modify autophagy levels to enhance lipid and glucose metabolism in diabetes [289].

Table 1 FDA-approved medicines with established indications on metabolism that could modulate autophagy

Metformin, adiponectin, leptin, the GLP1 agonists liraglutide and exenatide, oxyntomodulin (which is most likely a GLP1 agonist), the cannabinoid receptor antagonist rimonabant, and the histamine receptor agonist betahistine all have direct regulatory effects on autophagy [74, 85, 290]. Modulation of autophagy, on the other hand, is closely linked to a reduction in calorie intake. Furthermore, thiazolidinedione pioglitazone, an insulin sensitizer that acts in part by activating the PPAR, has been found to enhance autophagy [291, 292].

Other well-known autophagy promoters including trehalose, rapamycin, and imatinib are also employed in clinical trials to increase insulin sensitivity and cell function [293,294,295,296,297]. Imatinib, a tyrosine kinase inhibitor, lowers mitochondrial membrane potential (Δψm), increases ROS generation, and inhibits oxygen consumption and glycolysis while improving insulin sensitivity and promoting WAT browning through the inhibition of KIT, BCR-ABL, and platelet-derived growth factor receptor (PDGFR) [296, 298]. Imatinib, by inhibiting BCR–ABL activity, disrupts the BCR-ABL/PI3K/Akt/FOXO4/ATF5/mTOR pathway and as a result, may induce autophagy [297]. As a proteasome inhibitor, bortezomib reduces the activation of the mTOR pathway, which reduces ER stress, inflammation, and insulin resistance [299]. Although the FDA has approved a number of the aforementioned medications, the practicality of modifying autophagy has yet to be completely proven. Many medications that have metabolic benefits can modulate autophagy; however, autophagy modulation may not be their main/only mechanism of action.

Antidiabetic drugs have modulatory effects on the autophagy process. The cardioprotective effects of sitagliptin were mediated by the stimulation of autophagy [300]. Likewise, sitagliptin promoted autophagy through a method reliant on autophagy regulation, and decreased TG, low-density lipoprotein-cholesterol (LDL-c), and oxidative stress [300, 301]. Sitagliptin activated autophagy by stimulating AMPK and suppressing the mTOR signaling pathway, which in turn had positive effects on hepatic steatosis and insulin resistance [302]. Liraglutide administration by autophagy stimulation inhibited apoptosis and enhanced cognitive function and memory capacity through the suppression of mTOR and the expression of LC-3 II, Beclin-1, PI3K, AMPK, and Akt [300]. By suppressing liraglutide’s protective effects against glucose-induced cardiac injury, the use of 3BDO as an autophagy inhibitor made it clear that mTOR-dependent autophagy was at play [303]. Another antidiabetic drug, exenatide, has hepato-protective effects in diabetic mice, verified by enhanced autophagosome formation and upregulation of Beclin-1 and LC3A/B-II/I [300].

Conclusion

Given that previous investigations have been conducted on in vitro or animal models, more clinical studies should be performed to better understand the precise role of autophagy and its regulation in insulin resistance, as well as the crosstalk between autophagy and insulin function in varied tissues. Autophagy-targeted therapies provide promising possibilities for the treatment and prevention of insulin resistance. Many medications with good metabolic effects can modify autophagy, but affecting autophagy is not their only mechanism of action.

Availability of data and materials

Data will be made available on reasonable request.

References

  1. Brown JC, Harhay MO, Harhay MN. The value of anthropometric measures in nutrition and metabolism: comment on anthropometrically predicted visceral adipose tissue and blood-based biomarkers: a cross-sectional analysis. Nutr Metab Insights. 2019;12:1178638819831712.

    PubMed Central  Google Scholar 

  2. Galicia-Garcia U, Benito-Vicente A, Jebari S, Larrea-Sebal A, Siddiqi H, Uribe KB, et al. Pathophysiology of type 2 diabetes mellitus. Int J Mol Sci. 2020;21(17):6275.

    CAS  PubMed Central  Google Scholar 

  3. Petersen MC, Shulman GI. Mechanisms of insulin action and insulin resistance. Physiol Rev. 2018;98(4):2133–223.

    CAS  PubMed Central  Google Scholar 

  4. Rondinone CM, Wang L-M, Lonnroth P, Wesslau C, Pierce JH, Smith U. Insulin receptor substrate (IRS) 1 is reduced and IRS-2 is the main docking protein for phosphatidylinositol 3-kinase in adipocytes from subjects with non-insulin-dependent diabetes mellitus. Proc Natl Acad Sci. 1997;94(8):4171–5.

    CAS  PubMed Central  Google Scholar 

  5. Kim Y-B, Nikoulina SE, Ciaraldi TP, Henry RR, Kahn BB. Normal insulin-dependent activation of Akt/protein kinase B, with diminished activation of phosphoinositide 3-kinase, in muscle in type 2 diabetes. J Clin Investig. 1999;104(6):733–41.

    CAS  PubMed Central  Google Scholar 

  6. Ropelle ER, Flores MB, Cintra DE, Rocha GZ, Pauli JR, Morari J, et al. IL-6 and IL-10 anti-inflammatory activity links exercise to hypothalamic insulin and leptin sensitivity through IKKβ and ER stress inhibition. PLoS Biol. 2010;8(8): e1000465.

    PubMed Central  Google Scholar 

  7. Se S, Lee J, Goldfine AB. Inflammation and insulin resistance. J Clin Invest. 2006;116:1793–801.

    Google Scholar 

  8. Qatanani M, Lazar MA. Mechanisms of obesity-associated insulin resistance: many choices on the menu. Genes Dev. 2007;21(12):1443–55.

    CAS  Google Scholar 

  9. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11(5):373–84.

    CAS  PubMed  Google Scholar 

  10. Sah SP, Singh B, Choudhary S, Kumar A. Animal models of insulin resistance: a review. Pharmacol Rep. 2016;68(6):1165–77.

    CAS  Google Scholar 

  11. Fisher-Wellman KH, Neufer PD. Linking mitochondrial bioenergetics to insulin resistance via redox biology. Trends Endocrinol Metab. 2012;23(3):142–53.

    CAS  PubMed Central  Google Scholar 

  12. Cheng Z, Tseng Y, White MF. Insulin signaling meets mitochondria in metabolism. Trends Endocrinol Metab. 2010;21(10):589–98.

    CAS  PubMed Central  Google Scholar 

  13. Houstis N, Rosen ED, Lander ES. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature. 2006;440(7086):944–8.

    CAS  PubMed  Google Scholar 

  14. Evans JL, Maddux BA, Goldfine ID. The molecular basis for oxidative stress-induced insulin resistance. Antioxid Redox Signal. 2005;7(7–8):1040–52.

    CAS  Google Scholar 

  15. Hurrle S, Hsu WH. The etiology of oxidative stress in insulin resistance. Biomed J. 2017;40(5):257–62.

    PubMed Central  Google Scholar 

  16. Han J, Back SH, Hur J, Lin Y-H, Gildersleeve R, Shan J, et al. ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nat Cell Biol. 2013;15(5):481–90.

    CAS  PubMed Central  Google Scholar 

  17. Boden G, Duan X, Homko C, Molina EJ, Song W, Perez O, et al. Increase in endoplasmic reticulum stress–related proteins and genes in adipose tissue of obese, insulin-resistant individuals. Diabetes. 2008;57(9):2438–44.

    CAS  PubMed Central  Google Scholar 

  18. Lane JD, Gallagher LE, Chan EY. Early signalling events of autophagy. Essays Biochem. 2013;55:1–15.

    Google Scholar 

  19. Galluzzi L, Green DR. Autophagy-independent functions of the autophagy machinery. Cell. 2019;177(7):1682–99.

    CAS  PubMed Central  Google Scholar 

  20. Ohsumi Y. Historical landmarks of autophagy research. Cell Res. 2014;24(1):9–23.

    CAS  Google Scholar 

  21. Xie Z, Klionsky DJ. Autophagosome formation: core machinery and adaptations. Nat Cell Biol. 2007;9(10):1102–9.

    CAS  Google Scholar 

  22. Zachari M, Ganley IG. The mammalian ULK1 complex and autophagy initiation. Essays Biochem. 2017;61(6):585–96.

    PubMed Central  Google Scholar 

  23. Alers S, Löffler AS, Wesselborg S, Stork B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol Cell Biol. 2012;32(1):2–11.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Cao W, Li J, Yang K, Cao D. An overview of autophagy: mechanism, regulation and research progress. Bull Cancer. 2021;108:304.

    Google Scholar 

  25. Melia TJ, Lystad AH, Simonsen A. Autophagosome biogenesis: from membrane growth to closure. J Cell Biol. 2020;219(6).

  26. Yoshii SR, Mizushima N. Monitoring and measuring autophagy. Int J Mol Sci. 2017;18(9):1865.

    PubMed Central  Google Scholar 

  27. Mizushima N, Levine B. Autophagy in mammalian development and differentiation. Nat Cell Biol. 2010;12(9):823–30.

    CAS  PubMed Central  Google Scholar 

  28. Xie W, Zhou J. Aberrant regulation of autophagy in mammalian diseases. Biol Let. 2018;14(1):20170540.

    Google Scholar 

  29. RostamiRad A, Ebrahimi SSS, Sadeghi A, Taghikhani M, Meshkani R. Palmitate-induced impairment of autophagy turnover leads to increased apoptosis and inflammation in peripheral blood mononuclear cells. Immunobiology. 2018;223(3):269–78.

    CAS  Google Scholar 

  30. Alizadeh S, Mazloom H, Sadeghi A, Emamgholipour S, Golestani A, Noorbakhsh F, et al. Evidence for the link between defective autophagy and inflammation in peripheral blood mononuclear cells of type 2 diabetic patients. J Physiol Biochem. 2018;74(3):369–79.

    CAS  Google Scholar 

  31. Kovsan J, Blüher M, Tarnovscki T, Klöting N, Kirshtein B, Madar L, et al. Altered autophagy in human adipose tissues in obesity. J Clin Endocrinol Metab. 2011;96(2):E268–77.

    CAS  Google Scholar 

  32. Öst A, Svensson K, Ruishalme I, Brännmark C, Franck N, Krook H, et al. Attenuated mTOR signaling and enhanced autophagy in adipocytes from obese patients with type 2 diabetes. Mol Med. 2010;16(7):235–46.

    PubMed Central  Google Scholar 

  33. Clemente-Postigo M, Tinahones A, El Bekay R, Malagón MM, Tinahones FJ. The role of autophagy in white adipose tissue function: implications for metabolic health. Metabolites. 2020;10(5):179.

    CAS  PubMed Central  Google Scholar 

  34. Frendo-Cumbo S, Tokarz VL, Bilan PJ, Brumell JH, Klip A. Communication between autophagy and insulin action: at the crux of insulin action-insulin resistance? Front Cell Dev Biol. 2021;9.

  35. Soussi H, Clément K, Dugail I. Adipose tissue autophagy status in obesity: expression and flux—two faces of the picture. Autophagy. 2016;12(3):588–9.

    CAS  Google Scholar 

  36. Kovsan J, Blüher M, Tarnovscki T, Klöting N, Kirshtein B, Madar L, et al. Altered autophagy in human adipose tissues in obesity. Endocr Rev. 2010;31(6):945–6.

    Google Scholar 

  37. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124(3):471–84.

    CAS  Google Scholar 

  38. Jansen H, Van Essen P, Koenen T, Joosten L, Netea M, Tack C, et al. Autophagy activity is up-regulated in adipose tissue of obese individuals and modulates proinflammatory cytokine expression. Endocrinology. 2012;153(12):5866–74.

    CAS  Google Scholar 

  39. Codogno P, Meijer AJ. Autophagy: a potential link between obesity and insulin resistance. Cell Metab. 2010;11(6):449–51.

    CAS  Google Scholar 

  40. Jung CH, Jun CB, Ro S-H, Kim Y-M, Otto NM, Cao J, et al. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol Biol Cell. 2009;20(7):1992–2003.

    CAS  PubMed Central  Google Scholar 

  41. Hemmings BA, Restuccia DF. Pi3k-pkb/akt pathway. Cold Spring Harb Perspect Biol. 2012;4(9): a011189.

    PubMed Central  Google Scholar 

  42. Singh R, Xiang Y, Wang Y, Baikati K, Cuervo AM, Luu YK, et al. Autophagy regulates adipose mass and differentiation in mice. J Clin Investig. 2009;119(11):3329–39.

    CAS  PubMed Central  Google Scholar 

  43. Zhang Y, Goldman S, Baerga R, Zhao Y, Komatsu M, Jin S. Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice reveals a role in adipogenesis. Proc Natl Acad Sci. 2009;106(47):19860–5.

    CAS  PubMed Central  Google Scholar 

  44. Cai J, Pires KM, Ferhat M, Chaurasia B, Buffolo MA, Smalling R, et al. Autophagy ablation in adipocytes induces insulin resistance and reveals roles for lipid peroxide and Nrf2 signaling in adipose-liver crosstalk. Cell Rep. 2018;25(7):1708–17.

    CAS  PubMed Central  Google Scholar 

  45. Heilbronn LK, Campbell LV. Adipose tissue macrophages, low grade inflammation and insulin resistance in human obesity. Curr Pharm Des. 2008;14(12):1225–30.

    CAS  Google Scholar 

  46. Kang Y-H, Cho M-H, Kim J-Y, Kwon M-S, Peak J-J, Kang S-W, et al. Impaired macrophage autophagy induces systemic insulin resistance in obesity. Oncotarget. 2016;7(24):35577.

    PubMed Central  Google Scholar 

  47. Liu K, Zhao E, Ilyas G, Lalazar G, Lin Y, Haseeb M, et al. Impaired macrophage autophagy increases the immune response in obese mice by promoting proinflammatory macrophage polarization. Autophagy. 2015;11(2):271–84.

    CAS  PubMed Central  Google Scholar 

  48. Lavallard VJ, Meijer AJ, Codogno P, Gual P. Autophagy, signaling and obesity. Pharmacol Res. 2012;66(6):513–25.

    CAS  Google Scholar 

  49. Zhang Y, Zeng X, Jin S. Autophagy in adipose tissue biology. Pharmacol Res. 2012;66(6):505–12.

    CAS  Google Scholar 

  50. Zhang C, He Y, Okutsu M, Ong LC, Jin Y, Zheng L, et al. Autophagy is involved in adipogenic differentiation by repressing proteasome-dependent PPARγ2 degradation. Am J Physiol-Endocrinol Metab. 2013;305(4):E530–9.

    CAS  PubMed Central  Google Scholar 

  51. Baerga R, Zhang Y, Chen P-H, Goldman S, Jin SV. Targeted deletion of autophagy-related 5 (atg5) impairs adipogenesis in a cellular model and in mice. Autophagy. 2009;5(8):1118–30.

    CAS  Google Scholar 

  52. Dunlop EA, Tee AR. The kinase triad, AMPK, mTORC1 and ULK1, maintains energy and nutrient homoeostasis. Biochem Soc Trans. 2013;41(4):939–43.

    CAS  Google Scholar 

  53. Zhang X, Wu D, Wang C, Luo Y, Ding X, Yang X, et al. Sustained activation of autophagy suppresses adipocyte maturation via a lipolysis-dependent mechanism. Autophagy. 2020;16(9):1668–82.

    CAS  Google Scholar 

  54. Kim KH, Jeong YT, Oh H, Kim SH, Cho JM, Kim Y-N, et al. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat Med. 2013;19(1):83–92.

    CAS  Google Scholar 

  55. Ahmed B, Sultana R, Greene MW. Adipose tissue and insulin resistance in obese. Biomed Pharmacother. 2021;137: 111315.

    CAS  Google Scholar 

  56. McWilliams TG, Prescott AR, Montava-Garriga L, Ball G, Singh F, Barini E, et al. Basal mitophagy occurs independently of PINK1 in mouse tissues of high metabolic demand. Cell Metab. 2018;27(2):439–49.

    CAS  PubMed Central  Google Scholar 

  57. Pua HH, Guo J, Komatsu M, He Y-W. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J Immunol. 2009;182(7):4046–55.

    CAS  Google Scholar 

  58. Ashrafi G, Schwarz T. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ. 2013;20(1):31–42.

    CAS  PubMed  Google Scholar 

  59. Tsilingiris D, Tzeravini E, Koliaki C, Dalamaga M, Kokkinos A. The role of mitochondrial adaptation and metabolic flexibility in the pathophysiology of obesity and insulin resistance: an updated overview. Curr Obes Rep. 2021;10(3):191–213.

    Google Scholar 

  60. Maixner N, Kovsan J, Harman-Boehm I, Blüher M, Bashan N, Rudich A. Autophagy in adipose tissue. Obes Facts. 2012;5(5):710–21.

    Google Scholar 

  61. Yin J-J, Li Y-B, Wang Y, Liu G-D, Wang J, Zhu X-O, et al. The role of autophagy in endoplasmic reticulum stress-induced pancreatic β cell death. Autophagy. 2012;8(2):158–64.

    CAS  Google Scholar 

  62. Li H, Zhou B, Xu L, Liu J, Zang W, Wu S, et al. The reciprocal interaction between autophagic dysfunction and ER stress in adipose insulin resistance. Cell Cycle. 2014;13(4):565–79.

    Google Scholar 

  63. Behl T, Sehgal A, Bala R, Chadha S. Understanding the molecular mechanisms and role of autophagy in obesity. Mol Biol Rep. 2021;48:2881.

    CAS  Google Scholar 

  64. Zhou L, Zhang J, Fang Q, Liu M, Liu X, Jia W, et al. Autophagy-mediated insulin receptor down-regulation contributes to endoplasmic reticulum stress-induced insulin resistance. Mol Pharmacol. 2009;76(3):596–603.

    CAS  PubMed Central  Google Scholar 

  65. Zhang N, Cao M-M, Liu H, Xie G-Y, Li Y-B. Autophagy regulates insulin resistance following endoplasmic reticulum stress in diabetes. J Physiol Biochem. 2015;71(2):319–27.

    CAS  Google Scholar 

  66. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 2010;140(6):900–17.

    CAS  PubMed Central  Google Scholar 

  67. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.

    CAS  PubMed  Google Scholar 

  68. Ferhat M, Funai K, Boudina S. Autophagy in adipose tissue physiology and pathophysiology. Antioxid Redox Signal. 2019;31(6):487–501.

    CAS  PubMed Central  Google Scholar 

  69. Lim Y-M, Lim H, Hur KY, Quan W, Lee H-Y, Cheon H, et al. Systemic autophagy insufficiency compromises adaptation to metabolic stress and facilitates progression from obesity to diabetes. Nat Commun. 2014;5(1):1–14.

    Google Scholar 

  70. Nakahira K, Haspel JA, Rathinam VA, Lee S-J, Dolinay T, Lam HC, et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12(3):222–30.

    CAS  Google Scholar 

  71. Harris J, Hartman M, Roche C, Zeng SG, O’Shea A, Sharp FA, et al. Autophagy controls IL-1β secretion by targeting pro-IL-1β for degradation. J Biol Chem. 2011;286(11):9587–97.

    CAS  PubMed Central  Google Scholar 

  72. Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469(7329):221–5.

    CAS  PubMed  Google Scholar 

  73. Jung HS, Lee MS. Role of autophagy in diabetes and mitochondria. Ann N Y Acad Sci. 2010;1201(1):79–83.

    CAS  Google Scholar 

  74. Gan L, Liu Z, Luo D, Ren Q, Wu H, Li C, et al. Reduced endoplasmic reticulum stress-mediated autophagy is required for leptin alleviating inflammation in adipose tissue. Front Immunol. 2017;8:1507.

    PubMed Central  Google Scholar 

  75. Javaid HMA, Ko E, Joo EJ, Kwon SH, Park J-H, Shin S, et al. TNFα-induced NLRP3 inflammasome mediates adipocyte dysfunction and activates macrophages through adipocyte-derived lipocalin 2. Metabolism. 2023;142: 155527.

    CAS  Google Scholar 

  76. Cambi MPC, Baretta GAP, Magro DDO, Boguszewski CL, Ribeiro IB, Jirapinyo P, et al. Multidisciplinary approach for weight regain—how to manage this challenging condition: an expert review. Obes Surg. 2021;31(3):1290–303.

    Google Scholar 

  77. Zhang Y, Sowers JR, Ren J. Targeting autophagy in obesity: from pathophysiology to management. Nat Rev Endocrinol. 2018;14(6):356–76.

    CAS  Google Scholar 

  78. Alrushud AS, Rushton AB, Kanavaki AM, Greig CA. Effect of physical activity and dietary restriction interventions on weight loss and the musculoskeletal function of overweight and obese older adults with knee osteoarthritis: a systematic review and mixed method data synthesis. BMJ Open. 2017;7(6): e014537.

    PubMed Central  Google Scholar 

  79. Webb VL, Wadden TA. Intensive lifestyle intervention for obesity: principles, practices, and results. Gastroenterology. 2017;152(7):1752–64.

    Google Scholar 

  80. Halling JF, Pilegaard H. Autophagy-dependent beneficial effects of exercise. Cold Spring Harb Perspect Med. 2017;7(8): a029777.

    PubMed Central  Google Scholar 

  81. Kim KH, Lee M-S. Autophagy—a key player in cellular and body metabolism. Nat Rev Endocrinol. 2014;10(6):322–37.

    CAS  Google Scholar 

  82. He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, et al. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature. 2012;481(7382):511–5.

    CAS  PubMed Central  Google Scholar 

  83. Han X, Turdi S, Hu N, Guo R, Zhang Y, Ren J. Influence of long-term caloric restriction on myocardial and cardiomyocyte contractile function and autophagy in mice. J Nutr Biochem. 2012;23(12):1592–9.

    CAS  PubMed Central  Google Scholar 

  84. Jiang Y, Huang W, Wang J, Xu Z, He J, Lin X, et al. Metformin plays a dual role in MIN6 pancreatic β cell function through AMPK-dependent autophagy. Int J Biol Sci. 2014;10(3):268.

    PubMed Central  Google Scholar 

  85. Hussain Z, Khan JA. Food intake regulation by leptin: Mechanisms mediating gluconeogenesis and energy expenditure. Asian Pac J Trop Med. 2017;10(10):940–4.

    CAS  Google Scholar 

  86. Liu Y, Palanivel R, Rai E, Park M, Gabor TV, Scheid MP, et al. Adiponectin stimulates autophagy and reduces oxidative stress to enhance insulin sensitivity during high-fat diet feeding in mice. Diabetes. 2015;64(1):36–48.

    CAS  PubMed  Google Scholar 

  87. Sun F, Chai S, Li L, Yu K, Yang Z, Wu S, et al. Effects of glucagon-like peptide-1 receptor agonists on weight loss in patients with type 2 diabetes: a systematic review and network meta-analysis. J Diabetes Res. 2015;2015.

  88. Tomlinson B, Hu M, Zhang Y, Chan P, Liu Z-M. Investigational glucagon-like peptide-1 agonists for the treatment of obesity. Expert Opin Investig Drugs. 2016;25(10):1167–79.

    CAS  Google Scholar 

  89. Nowrouzi-Sohrabi P, Rezaei S, Jalali M, Ashourpour M, Ahmadipour A, Keshavarz P, et al. The effects of glucagon-like peptide-1 receptor agonists on glycemic control and anthropometric profiles among diabetic patients with non-alcoholic fatty liver disease: a systematic review and meta-analysis of randomized controlled trials. Eur J Pharmacol. 2021;893: 173823.

    CAS  Google Scholar 

  90. Courtney H, Nayar R, Rajeswaran C, Jandhyala R. Long-term management of type 2 diabetes with glucagon-like peptide-1 receptor agonists. Diabetes Metab Syndr Obes Targets Ther. 2017;10:79.

    CAS  Google Scholar 

  91. Newman JC, Covarrubias AJ, Zhao M, Yu X, Gut P, Ng C-P, et al. Ketogenic diet reduces midlife mortality and improves memory in aging mice. Cell Metab. 2017;26(3):547–57.

    CAS  PubMed Central  Google Scholar 

  92. Roberts MN, Wallace MA, Tomilov AA, Zhou Z, Marcotte GR, Tran D, et al. A ketogenic diet extends longevity and healthspan in adult mice. Cell Metab. 2017;26(3):539–46.

    CAS  PubMed Central  Google Scholar 

  93. Yamamoto S, Kuramoto K, Wang N, Situ X, Priyadarshini M, Zhang W, et al. Autophagy differentially regulates insulin production and insulin sensitivity. Cell Rep. 2018;23(11):3286–99.

    CAS  PubMed Central  Google Scholar 

  94. Kim O-K, Jun W, Lee J. Mechanism of ER stress and inflammation for hepatic insulin resistance in obesity. Ann Nutr Metab. 2015;67(4):218–27.

    CAS  Google Scholar 

  95. Khambu B, Yan S, Huda N, Liu G, Yin X-M, editors. Homeostatic role of autophagy in hepatocytes. Seminars in liver disease; 2018: Thieme Medical Publishers.

  96. Forbes JM. The physiological deadlock between AMPK and gluconeogenesis: SOGA, a novel protein, may provide the key. Am J Pathol. 2010;177(4):1600–2.

    PubMed Central  Google Scholar 

  97. Codogno P, Meijer AJ. Autophagy in the liver. J Hepatol. 2013;59(2):389–91.

    Google Scholar 

  98. Rui L. Energy metabolism in the liver. Compr Physiol. 2014;4(1):177.

    PubMed  PubMed Central  Google Scholar 

  99. Karsli-Uzunbas G, Guo JY, Price S, Teng X, Laddha SV, Khor S, et al. Autophagy is required for glucose homeostasis and lung tumor maintenance. Cancer Discov. 2014;4(8):914–27.

    CAS  PubMed Central  Google Scholar 

  100. Yang L, Li P, Fu S, Calay ES, Hotamisligil GS. Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Cell Metab. 2010;11(6):467–78.

    CAS  PubMed Central  Google Scholar 

  101. Liu H-Y, Han J, Cao SY, Hong T, Zhuo D, Shi J, et al. Hepatic autophagy is suppressed in the presence of insulin resistance and hyperinsulinemia: inhibition of FoxO1-dependent expression of key autophagy genes by insulin. J Biol Chem. 2009;284(45):31484–92.

    CAS  PubMed Central  Google Scholar 

  102. Ezaki J, Matsumoto N, Takeda-Ezaki M, Komatsu M, Takahashi K, Hiraoka Y, et al. Liver autophagy contributes to the maintenance of blood glucose and amino acid levels. Autophagy. 2011;7(7):727–36.

    CAS  PubMed Central  Google Scholar 

  103. den Hartigh LJ, Goodspeed L, Wang SA, Kenerson HL, Omer M, O’Brien KD, et al. Chronic oral rapamycin decreases adiposity, hepatic triglycerides and insulin resistance in male mice fed a diet high in sucrose and saturated fat. Exp Physiol. 2018;103(11):1469–80.

    Google Scholar 

  104. Cruces-Sande M, Arcones AC, Vila-Bedmar R, Val-Blasco A, Sharabi K, Díaz-Rodríguez D, et al. Autophagy mediates hepatic GRK2 degradation to facilitate glucagon-induced metabolic adaptation to fasting. FASEB J. 2020;34(1):399–409.

    CAS  Google Scholar 

  105. Jiang S, Young JL, Wang K, Qian Y, Cai L. Diabetic-induced alterations in hepatic glucose and lipid metabolism: the role of type 1 and type 2 diabetes mellitus. Mol Med Rep. 2020;22(2):603–11.

    CAS  PubMed Central  Google Scholar 

  106. Ke P-Y. Diverse functions of autophagy in liver physiology and liver diseases. Int J Mol Sci. 2019;20(2):300.

    PubMed Central  Google Scholar 

  107. Kanasaki K, Kawakita E, Koya D. Relevance of autophagy induction by gastrointestinal hormones: focus on the incretin-based drug target and glucagon. Front Pharmacol. 2019;10:476.

    CAS  PubMed Central  Google Scholar 

  108. Galsgaard KD, Lee J, Hubbard B, Zhang X-M, Cline G, Nasiri A, et al., editors. Glucagon promotes hepatic autophagy by AMPK-mediated mTORC1 inhibition. Diabetologia; 2020: Springer One New York Plaza, Suite 4600, New York, NY, United States.

  109. de Toro-Martín J, Fernández-Marcelo T, González-Rodríguez Á, Escrivá F, Valverde ÁM, Álvarez C, et al. Defective liver glycogen autophagy related to hyperinsulinemia in intrauterine growth-restricted newborn wistar rats. Sci Rep. 2020;10(1):1–14.

    Google Scholar 

  110. Han A, Zhang YN, Boehringer AS, Montes V, Andre MP, Erdman JW Jr, et al. Assessment of hepatic steatosis in nonalcoholic fatty liver disease by using quantitative US. Radiology. 2020;295(1):106–13.

    Google Scholar 

  111. Yu J, Marsh S, Hu J, Feng W, Wu C. The pathogenesis of nonalcoholic fatty liver disease: interplay between diet, gut microbiota, and genetic background. Gastroenterol Res Pract. 2016;2016.

  112. Nassir F, Rector RS, Hammoud GM, Ibdah JA. Pathogenesis and prevention of hepatic steatosis. Gastroenterol Hepatol. 2015;11(3):167.

    Google Scholar 

  113. Utzschneider KM, Kahn SE. The role of insulin resistance in nonalcoholic fatty liver disease. J Clin Endocrinol Metab. 2006;91(12):4753–61.

    CAS  Google Scholar 

  114. Allaire M, Rautou P-E, Codogno P, Lotersztajn S. Autophagy in liver diseases: time for translation? J Hepatol. 2019;70(5):985–98.

    Google Scholar 

  115. Wang K. Molecular mechanism of hepatic steatosis: pathophysiological role of autophagy. Expert Rev Mol Med. 2016;18.

  116. Kouroumalis E, Voumvouraki A, Augoustaki A, Samonakis DN. Autophagy in liver diseases. World J Hepatol. 2021;13(1):6.

    PubMed Central  Google Scholar 

  117. Grefhorst A, van de Peppel IP, Larsen LE, Jonker JW, Holleboom AG. The role of lipophagy in the development and treatment of non-alcoholic fatty liver disease. Front Endocrinol. 2021:1099.

  118. Zheng W, Zhou J, Song S, Kong W, Xia W, Chen L, et al. Dipeptidyl-peptidase 4 inhibitor sitagliptin ameliorates hepatic insulin resistance by modulating inflammation and autophagy in ob/ob mice. Int J Endocrinol. 2018;2018.

  119. El-Zayadi A-R. Hepatic steatosis: a benign disease or a silent killer. World J Gastroenterol: WJG. 2008;14(26):4120.

    PubMed Central  Google Scholar 

  120. Czaja MJ. Function of autophagy in nonalcoholic fatty liver disease. Dig Dis Sci. 2016;61(5):1304–13.

    CAS  PubMed Central  Google Scholar 

  121. Hazari Y, Bravo-San Pedro JM, Hetz C, Galluzzi L, Kroemer G. Autophagy in hepatic adaptation to stress. J Hepatol. 2020;72(1):183–96.

    CAS  Google Scholar 

  122. Xiong X, Tao R, DePinho RA, Dong XC. The autophagy-related gene 14 (Atg14) is regulated by forkhead box O transcription factors and circadian rhythms and plays a critical role in hepatic autophagy and lipid metabolism. J Biol Chem. 2012;287(46):39107–14.

    CAS  PubMed Central  Google Scholar 

  123. Hammoutene A, Lasselin J, Vion A-C, Colnot N, Paradis V, Lotersztajn S, et al. Defective autophagy in liver sinusoidal endothelial cells promotes non alcoholic steatohepatitis and fibrosis development. J Hepatol. 2018;68:S29.

    Google Scholar 

  124. Kwanten WJ, Martinet W, Francque SM. Autophagy in non-alcoholic fatty liver disease (NAFLD). Autophagy Curr Trends Cell Physiol Pathol. 2016:456–83.

  125. Xie X, Yan D, Li H, Zhu Q, Li J, Fang Y-p, et al. Enhancement of adiponectin ameliorates nonalcoholic fatty liver disease via inhibition of FOXO1 in type I diabetic rats. J Diabetes Res. 2018;2018.

  126. Cheng Z. The FoxO–autophagy Axis in health and disease. Trends Endocrinol Metab. 2019;30(9):658–71.

    CAS  Google Scholar 

  127. Dong XC. FOXO transcription factors in non-alcoholic fatty liver disease. Liver Res. 2017;1(3):168–73.

    PubMed Central  Google Scholar 

  128. Kwanten WJ, Martinet W, Michielsen PP, Francque SM. Role of autophagy in the pathophysiology of nonalcoholic fatty liver disease: a controversial issue. World J Gastroenterol: WJG. 2014;20(23):7325.

    PubMed Central  Google Scholar 

  129. Settembre C, Fraldi A, Medina DL, Ballabio A. Signals from the lysosome: a control centre for cellular clearance and energy metabolism. Nat Rev Mol Cell Biol. 2013;14(5):283–96.

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Di Malta C, Cinque L, Settembre C. Transcriptional regulation of autophagy: mechanisms and diseases. Front Cell Dev Biol. 2019;7:114.

    PubMed Central  Google Scholar 

  131. Zhang D, Ma Y, Liu J, Deng Y, Zhou B, Wen Y, et al. Metformin alleviates hepatic steatosis and insulin resistance in a mouse model of high-fat diet-induced nonalcoholic fatty liver disease by promoting transcription factor EB-dependent autophagy. Front Pharmacol. 2021:1907.

  132. Viollet B, Guigas B, Garcia NS, Leclerc J, Foretz M, Andreelli F. Cellular and molecular mechanisms of metformin: an overview. Clin Sci. 2012;122(6):253–70.

    CAS  Google Scholar 

  133. Settembre C, De Cegli R, Mansueto G, Saha PK, Vetrini F, Visvikis O, et al. TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat Cell Biol. 2013;15(6):647–58.

    CAS  PubMed Central  Google Scholar 

  134. Li B, Wu X, Chen H, Zhuang C, Zhang Z, Yao S, et al. miR199a-5p inhibits hepatic insulin sensitivity via suppression of ATG14-mediated autophagy. Cell Death Dis. 2018;9(3):1–15.

    Google Scholar 

  135. Ghareghani P, Shanaki M, Ahmadi S, Khoshdel AR, Rezvan N, Meshkani R, et al. Aerobic endurance training improves nonalcoholic fatty liver disease (NAFLD) features via miR-33 dependent autophagy induction in high fat diet fed mice. Obes Res Clin Pract. 2018;12(1):80–9.

    Google Scholar 

  136. Naito T, Kuma A, Mizushima N. Differential contribution of insulin and amino acids to the mTORC1-autophagy pathway in the liver and muscle. J Biol Chem. 2013;288(29):21074–81.

    CAS  PubMed Central  Google Scholar 

  137. Zhou W, Ye S. Rapamycin improves insulin resistance and hepatic steatosis in type 2 diabetes rats through activation of autophagy. Cell Biol Int. 2018;42(10):1282–91.

    CAS  Google Scholar 

  138. Ma D, Molusky MM, Song J, Hu C-R, Fang F, Rui C, et al. Autophagy deficiency by hepatic FIP200 deletion uncouples steatosis from liver injury in NAFLD. Mol Endocrinol. 2013;27(10):1643–54.

    CAS  PubMed Central  Google Scholar 

  139. Pereira S, Park E, Mori Y, Haber CA, Han P, Uchida T, et al. FFA-induced hepatic insulin resistance in vivo is mediated by PKCδ, NADPH oxidase, and oxidative stress. Am J Physiol-Endocrinol Metab. 2014;307(1):E34–46.

    CAS  PubMed Central  Google Scholar 

  140. Meshkani R, Adeli K. Hepatic insulin resistance, metabolic syndrome and cardiovascular disease. Clin Biochem. 2009;42(13–14):1331–46.

    CAS  Google Scholar 

  141. Tan SH, Shui G, Zhou J, Li JJE, Bay B-H, Wenk MR, et al. Induction of autophagy by palmitic acid via protein kinase C-mediated signaling pathway independent of mTOR (mammalian target of rapamycin). J Biol Chem. 2012;287(18):14364–76.

    CAS  PubMed Central  Google Scholar 

  142. Mei S, Ni H-M, Manley S, Bockus A, Kassel KM, Luyendyk JP, et al. Differential roles of unsaturated and saturated fatty acids on autophagy and apoptosis in hepatocytes. J Pharmacol Exp Ther. 2011;339(2):487–98.

    CAS  PubMed Central  Google Scholar 

  143. Abel ED, O’Shea KM, Ramasamy R. Insulin resistance: metabolic mechanisms and consequences in the heart. Arterioscler Thromb Vasc Biol. 2012;32(9):2068–76.

    CAS  PubMed Central  Google Scholar 

  144. Saito T, Kuma A, Sugiura Y, Ichimura Y, Obata M, Kitamura H, et al. Autophagy regulates lipid metabolism through selective turnover of NCoR1. Nat Commun. 2019;10(1):1–16.

    Google Scholar 

  145. Wang Y, Ding Y, Li J, Chavan H, Matye D, Ni H-M, et al. Targeting the enterohepatic bile acid signaling induces hepatic autophagy via a CYP7A1–AKT–mTOR axis in mice. Cell Mol Gastroenterol Hepatol. 2017;3(2):245–60.

    Google Scholar 

  146. Schulze RJ, Drižytė K, Casey CA, McNiven MA. Hepatic lipophagy: new insights into autophagic catabolism of lipid droplets in the liver. Hepatol Commun. 2017;1(5):359–69.

    PubMed Central  Google Scholar 

  147. Byrnes K, Blessinger S, Bailey N, Scaife R, Liu G, Khambu B. Therapeutic regulation of autophagy in hepatic metabolism. Acta Pharmaceutica Sinica B. 2021;12:33.

    PubMed Central  Google Scholar 

  148. Ohsaki Y, Cheng J, Fujita A, Tokumoto T, Fujimoto T. Cytoplasmic lipid droplets are sites of convergence of proteasomal and autophagic degradation of apolipoprotein B. Mol Biol Cell. 2006;17(6):2674–83.

    CAS  PubMed Central  Google Scholar 

  149. Zamani M, Taher J, Adeli K. Complex role of autophagy in regulation of hepatic lipid and lipoprotein metabolism. J Biomed Res. 2017;31(5):377.

    CAS  PubMed Central  Google Scholar 

  150. Fisher EA. The degradation of apolipoprotein B100: multiple opportunities to regulate VLDL triglyceride production by different proteolytic pathways. Biochimica et Biophysica Acta (BBA)-Mol Cell Biol Lipids. 2012;1821(5):778–81.

    CAS  Google Scholar 

  151. Andreo U, Guo L, Chirieac DV, Tuyama AC, Montenont E, Brodsky JL, et al. Insulin-stimulated degradation of apolipoprotein B100: roles of class II phosphatidylinositol-3-kinase and autophagy. PLoS ONE. 2013;8(3): e57590.

    CAS  PubMed Central  Google Scholar 

  152. Huang M, Yang X, Wang Z, Long J, Wang A, Zhang Y, et al. Lipophagy: a new perspective of natural products in type 2 diabetes mellitus treatment. Diabetes Metab Syndr Obes Targets Ther. 2021;14:2985.

    Google Scholar 

  153. Dimitriadis G, Mitrou P, Lambadiari V, Maratou E, Raptis SA. Insulin effects in muscle and adipose tissue. Diabetes Res Clin Pract. 2011;93:S52–9.

    CAS  Google Scholar 

  154. Álvarez-Mercado AI, Rojano-Alfonso C, Micó-Carnero M, Caballeria-Casals A, Uroz CP, Casillas-Ramírez A. New insights into the role of autophagy in liver surgery in the setting of metabolic syndrome and related diseases. Front Cell Dev Biol. 2021;9:1250.

    Google Scholar 

  155. Özcan U, Cao Q, Yilmaz E, Lee A-H, Iwakoshi NN, Özdelen E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004;306(5695):457–61.

    Google Scholar 

  156. Villalobos-Labra R, Subiabre M, Toledo F, Pardo F, Sobrevia L. Endoplasmic reticulum stress and development of insulin resistance in adipose, skeletal, liver, and foetoplacental tissue in diabesity. Mol Aspects Med. 2019;66:49–61.

    CAS  Google Scholar 

  157. Ohoka N, Yoshii S, Hattori T, Onozaki K, Hayashi H. TRB3, a novel ER stress-inducible gene, is induced via ATF4–CHOP pathway and is involved in cell death. EMBO J. 2005;24(6):1243–55.

    CAS  PubMed Central  Google Scholar 

  158. Akhter MS, Uddin MA, Kubra K-T, Barabutis N. Autophagy, unfolded protein response and lung disease. Curr Res Cell Biol. 2020:100003.

  159. Ding W-X, Ni H-M, Gao W, Yoshimori T, Stolz DB, Ron D, et al. Linking of autophagy to ubiquitin-proteasome system is important for the regulation of endoplasmic reticulum stress and cell viability. Am J Pathol. 2007;171(2):513–24.

    CAS  PubMed Central  Google Scholar 

  160. Yorimitsu T, Nair U, Yang Z, Klionsky DJ. Endoplasmic reticulum stress triggers autophagy. J Biol Chem. 2006;281(40):30299–304.

    CAS  Google Scholar 

  161. Ogata M, Hino S-i, Saito A, Morikawa K, Kondo S, Kanemoto S, et al. Autophagy is activated for cell survival after endoplasmic ReticulumStress. Mol Cell Biol. 2006;26(24):9220–31.

    CAS  PubMed Central  Google Scholar 

  162. Ding W-X, Ni H-M, Gao W, Hou Y-F, Melan MA, Chen X, et al. Differential effects of endoplasmic reticulum stress-induced autophagy on cell survival. J Biol Chem. 2007;282(7):4702–10.

    CAS  Google Scholar 

  163. Ding W-X. Role of autophagy in liver physiology and pathophysiology. World J Biol Chem. 2010;1(1):3.

    PubMed Central  Google Scholar 

  164. Yan H, Gao Y, Zhang Y. Inhibition of JNK suppresses autophagy and attenuates insulin resistance in a rat model of nonalcoholic fatty liver disease. Mol Med Rep. 2017;15(1):180–6.

    CAS  Google Scholar 

  165. Wang H, Sun R-Q, Zeng X-Y, Zhou X, Li S, Jo E, et al. Restoration of autophagy alleviates hepatic ER stress and impaired insulin signalling transduction in high fructose-fed male mice. Endocrinology. 2015;156(1):169–81.

    Google Scholar 

  166. Gonzalez-Rodriguez A, Mayoral R, Agra N, Valdecantos M, Pardo V, Miquilena-Colina M, et al. Impaired autophagic flux is associated with increased endoplasmic reticulum stress during the development of NAFLD. Cell Death Dis. 2014;5(4):e1179.

    CAS  PubMed Central  Google Scholar 

  167. Ma X, McKeen T, Zhang J, Ding W-X. Role and mechanisms of mitophagy in liver diseases. Cells. 2020;9(4):837.

    CAS  PubMed Central  Google Scholar 

  168. Doblado L, Lueck C, Rey C, Samhan-Arias AK, Prieto I, Stacchiotti A, et al. Mitophagy in human diseases. Int J Mol Sci. 2021;22(8):3903.

    CAS  PubMed Central  Google Scholar 

  169. Rocha M, Apostolova N, Diaz-Rua R, Muntane J, Victor VM. Mitochondria and T2D: role of autophagy, ER stress, and inflammasome. Trends Endocrinol Metab. 2020;31:725.

    CAS  Google Scholar 

  170. Tiganis T. Reactive oxygen species and insulin resistance: the good, the bad and the ugly. Trends Pharmacol Sci. 2011;32(2):82–9.

    CAS  Google Scholar 

  171. Besse-Patin A, Estall JL. An intimate relationship between ROS and insulin signalling: implications for antioxidant treatment of fatty liver disease. Int J Cell Biol. 2014;2014.

  172. Nakamura S, Takamura T, Matsuzawa-Nagata N, Takayama H, Misu H, Noda H, et al. Palmitate induces insulin resistance in H4IIEC3 hepatocytes through reactive oxygen species produced by mitochondria. J Biol Chem. 2009;284(22):14809–18.

    CAS  PubMed Central  Google Scholar 

  173. Su Z, Nie Y, Huang X, Zhu Y, Feng B, Tang L, et al. Mitophagy in hepatic insulin resistance: therapeutic potential and concerns. Front Pharmacol. 2019;10:1193.

    CAS  PubMed Central  Google Scholar 

  174. Feng J, Lu S, Ou B, Liu Q, Dai J, Ji C, et al. The role of JNk signaling pathway in obesity-driven insulin resistance. Diabetes Metab Syndr Obes Targets Ther. 2020;13:1399.

    CAS  Google Scholar 

  175. Pietrocola F, Pedro B-S, Manuel J. Targeting autophagy to counteract obesity-associated oxidative stress. Antioxidants. 2021;10(1):102.

    CAS  PubMed Central  Google Scholar 

  176. Muriach M, Flores-Bellver M, Romero FJ, Barcia JM. Diabetes and the brain: oxidative stress, inflammation, and autophagy. Oxid Med Cell Longevity. 2014;2014.

  177. Crişan TO, Plantinga TS, van de Veerdonk FL, Farcaş MF, Stoffels M, Kullberg B-J, et al. Inflammasome-independent modulation of cytokine response by autophagy in human cells. PLoS ONE. 2011;6(4): e18666.

    PubMed Central  Google Scholar 

  178. Condello M, Pellegrini E, Caraglia M, Meschini S. Targeting autophagy to overcome human diseases. Int J Mol Sci. 2019;20(3):725.

    CAS  PubMed Central  Google Scholar 

  179. Barbier L, Ferhat M, Salamé E, Robin A, Herbelin A, Gombert J-M, et al. Interleukin-1 family cytokines: keystones in liver inflammatory diseases. Front Immunol. 2019;10:2014.

    CAS  PubMed Central  Google Scholar 

  180. Mao Y, Yu F, Wang J, Guo C, Fan X. Autophagy: a new target for nonalcoholic fatty liver disease therapy. Hepatic Med Evid Res. 2016;8:27.

    Google Scholar 

  181. Zeng T-s, Liu F-m, Zhou J, Pan S-x, Xia W-f, Chen L-l. Depletion of Kupffer cells attenuates systemic insulin resistance, inflammation and improves liver autophagy in high-fat diet fed mice. Endocrine J. 2015:EJ15-0046.

  182. Neel BA, Lin Y, Pessin JE. Skeletal muscle autophagy: a new metabolic regulator. Trends Endocrinol Metab. 2013;24(12):635–43.

    CAS  Google Scholar 

  183. Consitt LA, Dudley C, Saxena G. Impact of endurance and resistance training on skeletal muscle glucose metabolism in older adults. Nutrients. 2019;11(11):2636.

    CAS  PubMed Central  Google Scholar 

  184. Qin B, Zhou Z, He J, Yan C, Ding S. IL-6 inhibits starvation-induced autophagy via the STAT3/Bcl-2 signaling pathway. Sci Rep. 2015;5(1):1–10.

    Google Scholar 

  185. Song X, Shen Q, Fan L, Yu Q, Jia X, Sun Y, et al. Dehydroepiandrosterone-induced activation of mTORC1 and inhibition of autophagy contribute to skeletal muscle insulin resistance in a mouse model of polycystic ovary syndrome. Oncotarget. 2018;9(15):11905.

    PubMed Central  Google Scholar 

  186. Yang L, Lin H, Lin W, Xu X. Exercise ameliorates insulin resistance of type 2 diabetes through motivating short-chain fatty acid-mediated skeletal muscle cell autophagy. Biology. 2020;9(8):203.

    CAS  PubMed Central  Google Scholar 

  187. Shi L, Zhang T, Zhou Y, Zeng X, Ran L, Zhang Q, et al. Dihydromyricetin improves skeletal muscle insulin sensitivity by inducing autophagy via the AMPK-PGC-1α-Sirt3 signaling pathway. Endocrine. 2015;50(2):378–89.

    CAS  Google Scholar 

  188. Møller AB, Kampmann U, Hedegaard J, Thorsen K, Nordentoft I, Vendelbo MH, et al. Altered gene expression and repressed markers of autophagy in skeletal muscle of insulin resistant patients with type 2 diabetes. Sci Rep. 2017;7(1):1–11.

    Google Scholar 

  189. Kruse R, Vind BF, Petersson SJ, Kristensen JM, Højlund K. Markers of autophagy are adapted to hyperglycaemia in skeletal muscle in type 2 diabetes. Diabetologia. 2015;58(9):2087–95.

    CAS  Google Scholar 

  190. Sarparanta J, Garcia-Macia M, Singh R. Autophagy and mitochondria in obesity and type 2 diabetes. Curr Diabetes Rev. 2017;13(4):352–69.

    CAS  Google Scholar 

  191. Lv P, Huang J, Yang J, Deng Y, Xu J, Zhang X, et al. Autophagy in muscle of glucose-infusion hyperglycemia rats and streptozotocin-induced hyperglycemia rats via selective activation of m-TOR or FoxO3. PLoS ONE. 2014;9(2): e87254.

    PubMed Central  Google Scholar 

  192. Færch K, Vistisen D, Pacini G, Torekov SS, Johansen NB, Witte DR, et al. Insulin resistance is accompanied by increased fasting glucagon and delayed glucagon suppression in individuals with normal and impaired glucose regulation. Diabetes. 2016;65(11):3473–81.

    Google Scholar 

  193. Hegarty B, Furler S, Ye J, Cooney G, Kraegen E. The role of intramuscular lipid in insulin resistance. Acta Physiol Scand. 2003;178(4):373–83.

    CAS  Google Scholar 

  194. Gilbert M. Role of skeletal muscle lipids in the pathogenesis of insulin resistance of obesity and type 2 diabetes. J Diabetes Invest. 2021;12(11):1934–41.

    CAS  Google Scholar 

  195. Sadeghi A, Shabani M, Alizadeh S, Meshkani R. Interplay between oxidative stress and autophagy function and its role in inflammatory cytokine expression induced by palmitate in skeletal muscle cells. Cytokine. 2020;125: 154835.

    CAS  Google Scholar 

  196. Lam T, Harmancey R, Vasquez H, Gilbert B, Patel N, Hariharan V, et al. Reversal of intramyocellular lipid accumulation by lipophagy and a p62-mediated pathway. Cell Death Discovery. 2016;2(1):1–12.

    Google Scholar 

  197. da Silva Rosa SC, Martens MD, Field JT, Nguyen L, Kereliuk SM, Hai Y, et al. BNIP3L/Nix-induced mitochondrial fission, mitophagy, and impaired myocyte glucose uptake are abrogated by PRKA/PKA phosphorylation. Autophagy. 2021;17(9):2257–72.

    Google Scholar 

  198. Morales-Scholz MG, Swinton C, Murphy RM, Kowalski GM, Bruce CR, Howlett KF, et al. Autophagy is not involved in lipid accumulation and the development of insulin resistance in skeletal muscle. Biochem Biophys Res Commun. 2021;534:533–9.

    CAS  Google Scholar 

  199. Paré M, Baechler B, Fajardo V, Earl E, Wong E, Campbell T, et al. Effect of acute and chronic autophagy deficiency on skeletal muscle apoptotic signaling, morphology, and function. Biochimica et Biophysica Acta (BBA) Mol Cell Res. 2017;1864(4):708–18.

    Google Scholar 

  200. Campbell TL, Mitchell AS, McMillan EM, Bloemberg D, Pavlov D, Messa I, et al. High-fat feeding does not induce an autophagic or apoptotic phenotype in female rat skeletal muscle. Exp Biol Med. 2015;240(5):657–68.

    CAS  Google Scholar 

  201. Jani S, Da Eira D, Hadday I, Bikopoulos G, Mohasses A, de Pinho RA, et al. Distinct mechanisms involving diacylglycerol, ceramides, and inflammation underlie insulin resistance in oxidative and glycolytic muscles from high fat-fed rats. Sci Rep. 2021;11(1):1–12.

    Google Scholar 

  202. Cerella C, Dicato M, Diederich M. Modulatory roles of glycolytic enzymes in cell death. Biochem Pharmacol. 2014;92(1):22–30.

    CAS  Google Scholar 

  203. Roberts DJ, Tan-Sah VP, Ding EY, Smith JM, Miyamoto S. Hexokinase-II positively regulates glucose starvation-induced autophagy through TORC1 inhibition. Mol Cell. 2014;53(4):521–33.

    CAS  PubMed Central  Google Scholar 

  204. Nakai N, Kitai S, Iida N, Inoue S, Nakata K, Murakami T, et al. Induction of autophagy and changes in cellular metabolism in glucose starved C2C12 myotubes. J Nutr Sci Vitaminol. 2020;66(1):41–7.

    CAS  Google Scholar 

  205. Altajar S, Baffy G. Skeletal muscle dysfunction in the development and progression of nonalcoholic fatty liver disease. J Clin Transl Hepatol. 2020;8(4):414.

    PubMed Central  Google Scholar 

  206. McMillan EM, Paré M-F, Baechler BL, Graham DA, Rush JW, Quadrilatero J. Autophagic signaling and proteolytic enzyme activity in cardiac and skeletal muscle of spontaneously hypertensive rats following chronic aerobic exercise. PLoS ONE. 2015;10(3): e0119382.

    PubMed Central  Google Scholar 

  207. Ryu JY, Choi HM, Yang H-I, Kim KS. Dysregulated autophagy mediates sarcopenic obesity and its complications via AMPK and PGC1α signaling pathways: potential involvement of gut dysbiosis as a pathological link. Int J Mol Sci. 2020;21(18):6887.

    CAS  PubMed Central  Google Scholar 

  208. Chen W, Wang L, You W, Shan T. Myokines mediate the cross talk between skeletal muscle and other organs. J Cell Physiol. 2021;236(4):2393–412.

    CAS  Google Scholar 

  209. Amor M, Itariu BK, Moreno-Viedma V, Keindl M, Jürets A, Prager G, et al. Serum myostatin is upregulated in obesity and correlates with insulin resistance in humans. Exp Clin Endocrinol Diabetes. 2019;127(08):550–6.

    CAS  Google Scholar 

  210. Liu XH, Bauman WA, Cardozo CP. Myostatin inhibits glucose uptake via suppression of insulin-dependent and-independent signaling pathways in myoblasts. Physiol Rep. 2018;6(17): e13837.

    PubMed Central  Google Scholar 

  211. Pesce M, Ballerini P, Paolucci T, Puca I, Farzaei MH, Patruno A. Irisin and autophagy: first update. Int J Mol Sci. 2020;21(20):7587.

    CAS  PubMed Central  Google Scholar 

  212. Zhang C, McFarlane C, Lokireddy S, Bonala S, Ge X, Masuda S, et al. Myostatin-deficient mice exhibit reduced insulin resistance through activating the AMP-activated protein kinase signalling pathway. Diabetologia. 2011;54(6):1491–501.

    CAS  Google Scholar 

  213. Eilers W, Cleasby M, Foster K. Development of antisense-mediated myostatin knockdown for the treatment of insulin resistance. Sci Rep. 2021;11(1):1–10.

    Google Scholar 

  214. Camporez J-PG, Petersen MC, Abudukadier A, Moreira GV, Jurczak MJ, Friedman G, et al. Anti-myostatin antibody increases muscle mass and strength and improves insulin sensitivity in old mice. Proc Natl Acad Sci. 2016;113(8):2212–7.

    CAS  PubMed Central  Google Scholar 

  215. Ferraro E, Giammarioli AM, Chiandotto S, Spoletini I, Rosano G. Exercise-induced skeletal muscle remodeling and metabolic adaptation: redox signaling and role of autophagy. Antioxid Redox Signal. 2014;21(1):154–76.

    CAS  PubMed Central  Google Scholar 

  216. Ye X, Shen Y, Ni C, Ye J, Xin Y, Zhang W, et al. Irisin reverses insulin resistance in C2C12 cells via the p38-MAPK-PGC-1α pathway. Peptides. 2019;119: 170120.

    CAS  Google Scholar 

  217. Mi Q, Li Y, Wang M, Yang G, Zhao X, Liu H, et al. Circulating C1q/TNF-related protein isoform 15 is a marker for the presence of metabolic syndrome. Diabetes Metab Res Rev. 2019;35(1): e3085.

    Google Scholar 

  218. Shokoohi Nahrkhalaji A, Ahmadi R, Fadaei R, Panahi G, Razzaghi M, Fallah S. Higher serum level of CTRP15 in patients with coronary artery disease is associated with disease severity, body mass index and insulin resistance. Arch Physiol Biochem. 2022;128(1):276–80.

    CAS  Google Scholar 

  219. Mohassel Azadi S, Shateri H, Mohammadi M, Fadaei R, Sajedi F, Ziamajidi N. Increased circulating level of CTRP15 in patients with type 2 diabetes mellitus and its relation with inflammation and insulin resistance. J Diabetes Metab Disord. 2021;20(2):1499–504.

    CAS  PubMed Central  Google Scholar 

  220. Vatannejad A, Fadaei R, Salimi F, Fouani FZ, Habibi B, Shapourizadeh S, et al. Plasma Complement C1q/tumor necrosis factor-related protein 15 concentration is associated with polycystic ovary syndrome. PLoS ONE. 2022;17(6): e0263658.

    CAS  PubMed Central  Google Scholar 

  221. Pourranjbar M, Arabnejad N, Naderipour K, Rafie F. Effects of aerobic exercises on serum levels of myonectin and insulin resistance in obese and overweight women. J Med Life. 2018;11(4):381.

    PubMed Central  Google Scholar 

  222. Seldin MM, Lei X, Tan SY, Stanson KP, Wei Z, Wong GW. Skeletal muscle-derived myonectin activates the mammalian target of rapamycin (mTOR) pathway to suppress autophagy in liver. J Biol Chem. 2013;288(50):36073–82.

    CAS  PubMed Central  Google Scholar 

  223. Siewko K, Maciulewski R, Zielinska-Maciulewska A, Poplawska-Kita A, Szumowski P, Wawrusiewicz-Kurylonek N, et al. Interleukin-6 and interleukin-15 as possible biomarkers of the risk of autoimmune diabetes development. BioMed Res Int. 2019;2019:1.

    Google Scholar 

  224. Krolopp JE, Thornton SM, Abbott MJ. IL-15 activates the Jak3/STAT3 signaling pathway to mediate glucose uptake in skeletal muscle cells. Front Physiol. 2016;7:626.

    PubMed Central  Google Scholar 

  225. 藤本拓. Overexpression of interleukin-15 exhibits improved glucose tolerance and promotes GLUT4 translocation via AMP-activated protein kinase pathway in skeletal muscle: 大阪大学; 2019.

  226. Orsatti CL, Orsatti FL, Bezerra TG, Quevedo A, Nahas EAP. Interleukin-15 are associated with insulin resistance in postmenopausal women with metabolic syndrome. Gynecol Endocrinol. 2022;38(9):765–70.

    CAS  Google Scholar 

  227. Zhu L, Xie X, Zhang L, Wang H, Jie Z, Zhou X, et al. TBK-binding protein 1 regulates IL-15-induced autophagy and NKT cell survival. Nat Commun. 2018;9(1):1–14.

    Google Scholar 

  228. Ara A, Wu Z, Xu A, Ahmed KA, Leary SC, Islam MF, et al. The critical role of AMPKα1 in regulating autophagy and mitochondrial respiration in IL-15-stimulated mTORC1Weak signal-induced T cell memory: an interplay between Yin (AMPKα1) and Yang (mTORC1) energy sensors in T cell differentiation. Int J Mol Sci. 2022;23(17):9534.

    CAS  PubMed Central  Google Scholar 

  229. Yan J, Feng Z, Liu J, Shen W, Wang Y, Wertz K, et al. Enhanced autophagy plays a cardinal role in mitochondrial dysfunction in type 2 diabetic Goto-Kakizaki (GK) rats: ameliorating effects of (−)-epigallocatechin-3-gallate. J Nutr Biochem. 2012;23(7):716–24.

    CAS  Google Scholar 

  230. Ehrlicher SE, Stierwalt HD, Newsom SA, Robinson MM. Short-term high-fat feeding does not alter mitochondrial lipid respiratory capacity but triggers mitophagy response in skeletal muscle of mice. Front Endocrinol. 2021;12: 651211.

    Google Scholar 

  231. Haines MS, Dichtel LE, Santoso K, Torriani M, Miller KK, Bredella MA. Association between muscle mass and insulin sensitivity independent of detrimental adipose depots in young adults with overweight/obesity. Int J Obes. 2020;44(9):1851–8.

    CAS  Google Scholar 

  232. Dobrowolny G, Aucello M, Rizzuto E, Beccafico S, Mammucari C, Bonconpagni S, et al. Skeletal muscle is a primary target of SOD1G93A-mediated toxicity. Cell Metab. 2008;8(5):425–36.

    CAS  Google Scholar 

  233. Suzuki N, Motohashi N, Uezumi A, Fukada S-I, Yoshimura T, Itoyama Y, et al. NO production results in suspension-induced muscle atrophy through dislocation of neuronal NOS. J Clin Invest. 2007;117(9):2468–76.

    CAS  PubMed Central  Google Scholar 

  234. McClung JM, Judge AR, Powers SK, Yan Z. p38 MAPK links oxidative stress to autophagy-related gene expression in cachectic muscle wasting. Am J Physiol Cell Physiol. 2010;298(3):C542–9.

    CAS  Google Scholar 

  235. Malicdan MC, Noguchi S, Nonaka I, Saftig P, Nishino I. Lysosomal myopathies: an excessive build-up in autophagosomes is too much to handle. Neuromuscul Disord. 2008;18(7):521–9.

    Google Scholar 

  236. Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E, Komatsu M, et al. Autophagy is required to maintain muscle mass. Cell Metab. 2009;10(6):507–15.

    CAS  Google Scholar 

  237. Raben N, Hill V, Shea L, Takikita S, Baum R, Mizushima N, et al. Suppression of autophagy in skeletal muscle uncovers the accumulation of ubiquitinated proteins and their potential role in muscle damage in Pompe disease. Hum Mol Genet. 2008;17(24):3897–908.

    CAS  PubMed Central  Google Scholar 

  238. Qi Z, Chen L. Endoplasmic reticulum stress and autophagy. Autophagy Biol Dis. 2019:167–77.

  239. Madaro L, Marrocco V, Carnio S, Sandri M, Bouché M. Intracellular signaling in ER stress-induced autophagy in skeletal muscle cells. FASEB J. 2013;27(5):1990–2000.

    CAS  Google Scholar 

  240. Ijuin T, Hosooka T, Takenawa T. Phosphatidylinositol 3, 4, 5-trisphosphate phosphatase SKIP links endoplasmic reticulum stress in skeletal muscle to insulin resistance. Mol Cell Biol. 2016;36(1):108–18.

    CAS  Google Scholar 

  241. Ahlstrom P, Rai E, Chakma S, Cho HH, Rengasamy P, Sweeney G. Adiponectin improves insulin sensitivity via activation of autophagic flux. J Mol Endocrinol. 2017;59(4):339–50.

    CAS  Google Scholar 

  242. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Investig. 2003;112(12):1821–30.

    CAS  PubMed Central  Google Scholar 

  243. Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the link between insulin resistance, obesity and diabetes. Trends Immunol. 2004;25(1):4–7.

    CAS  Google Scholar 

  244. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J Clin Investig. 2006;116(7):1793–801.

    CAS  PubMed Central  Google Scholar 

  245. Solinas G, Becattini B. JNK at the crossroad of obesity, insulin resistance, and cell stress response. Molecular metabolism. 2017;6(2):174–84.

    CAS  Google Scholar 

  246. Tilg H, Moschen AR. Inflammatory mechanisms in the regulation of insulin resistance. Mol Med. 2008;14(3):222–31.

    CAS  PubMed Central  Google Scholar 

  247. Moon S, Jung HS. Endoplasmic reticulum stress and dysregulated autophagy in human pancreatic beta cells. Diabetes Metab J. 2022;46(4):533–42.

    PubMed Central  Google Scholar 

  248. Bartolome A, Guillen C, Benito M. Autophagy plays a protective role in endoplasmic reticulum stress-mediated pancreatic β cell death. Autophagy. 2012;8(12):1757–68.

    CAS  PubMed Central  Google Scholar 

  249. Ji J, Petropavlovskaia M, Khatchadourian A, Patapas J, Makhlin J, Rosenberg L, et al. Type 2 diabetes is associated with suppression of autophagy and lipid accumulation in β-cells. J Cell Mol Med. 2019;23(4):2890–900.

    CAS  PubMed Central  Google Scholar 

  250. Mir SU, George NM, Zahoor L, Harms R, Guinn Z, Sarvetnick NE. Inhibition of autophagic turnover in β-cells by fatty acids and glucose leads to apoptotic cell death. J Biol Chem. 2015;290(10):6071–85.

    CAS  Google Scholar 

  251. Elumalai S, Karunakaran U, Moon JS, Won KC. High glucose-induced PRDX3 acetylation contributes to glucotoxicity in pancreatic β-cells: prevention by Teneligliptin. Free Radical Biol Med. 2020;160:618–29.

    CAS  Google Scholar 

  252. Yun HR, Jo YH, Kim J, Shin Y, Kim SS, Choi TG. Roles of autophagy in oxidative stress. Int J Mol Sci. 2020;21(9):3289.

    CAS  PubMed Central  Google Scholar 

  253. Xia G, Zhu T, Li X, Jin Y, Zhou J, Xiao J. ROS-mediated autophagy through the AMPK signaling pathway protects INS-1 cells from human islet amyloid polypeptide-induced cytotoxicity. Mol Med Rep. 2018;18(3):2744–52.

    CAS  PubMed Central  Google Scholar 

  254. Shigihara N, Fukunaka A, Hara A, Komiya K, Honda A, Uchida T, et al. Human IAPP–induced pancreatic β cell toxicity and its regulation by autophagy. J Clin Investig. 2014;124(8):3634–44.

    CAS  PubMed Central  Google Scholar 

  255. Kim J, Park K, Kim MJ, Lim H, Kim KH, Kim S-W, et al. An autophagy enhancer ameliorates diabetes of human IAPP-transgenic mice through clearance of amyloidogenic oligomer. Nat Commun. 2021;12(1):183.

    CAS  PubMed Central  Google Scholar 

  256. Park MJ, Choi KM. Interplay of skeletal muscle and adipose tissue: sarcopenic obesity. Metabolism. 2023;144:155577.

    CAS  Google Scholar 

  257. Kosacka J, Kern M, Klöting N, Paeschke S, Rudich A, Haim Y, et al. Autophagy in adipose tissue of patients with obesity and type 2 diabetes. Mol Cell Endocrinol. 2015;409:21–32.

    CAS  Google Scholar 

  258. Iula L, Keitelman IA, Sabbione F, Fuentes F, Guzman M, Galletti JG, et al. Autophagy mediates interleukin-1β secretion in human neutrophils. Front Immunol. 2018;9:269.

    PubMed Central  Google Scholar 

  259. Saitoh T, Fujita N, Jang MH, Uematsu S, Yang B-G, Satoh T, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1β production. Nature. 2008;456(7219):264–8.

    CAS  Google Scholar 

  260. Kume S, Yamahara K, Yasuda M, Maegawa H, Koya D. Autophagy: emerging therapeutic target for diabetic nephropathy. Semin Nephrol. 2014;34(1):9–16.

    CAS  Google Scholar 

  261. Huang S-S, Ding D-F, Chen S, Dong C-L, Ye X-L, Yuan Y-G, et al. Resveratrol protects podocytes against apoptosis via stimulation of autophagy in a mouse model of diabetic nephropathy. Sci Rep. 2017;7(1):45692.

    CAS  PubMed Central  Google Scholar 

  262. Liu N, Xu L, Shi Y, Zhuang S. Podocyte autophagy: a potential therapeutic target to prevent the progression of diabetic nephropathy. J Diabetes Res. 2017;2017.

  263. Lin JS, Susztak K. Podocytes: the Weakest Link in Diabetic Kidney Disease? Curr Diab Rep. 2016;16(5):45.

    PubMed Central  Google Scholar 

  264. Deshpande S, Abdollahi M, Wang M, Lanting L, Kato M, Natarajan R. Reduced autophagy by a microRNA-mediated signaling cascade in diabetes-induced renal glomerular hypertrophy. Sci Rep. 2018;8(1):1–13.

    Google Scholar 

  265. Yang D, Livingston MJ, Liu Z, Dong G, Zhang M, Chen J-K, et al. Autophagy in diabetic kidney disease: regulation, pathological role and therapeutic potential. Cell Mol Life Sci. 2018;75(4):669–88.

    CAS  PubMed  Google Scholar 

  266. Yamahara K, Yasuda M, Kume S, Koya D, Maegawa H, Uzu T. The role of autophagy in the pathogenesis of diabetic nephropathy. J Diabetes Res. 2013;2013:1.

    Google Scholar 

  267. Kitada M, Ogura Y, Monno I, Koya D. Regulating autophagy as a therapeutic target for diabetic nephropathy. Curr Diab Rep. 2017;17(7):53.

    Google Scholar 

  268. Tang C, Livingston MJ, Liu Z, Dong Z. Autophagy in kidney homeostasis and disease. Nat Rev Nephrol. 2020;16(9):489–508.

    CAS  PubMed Central  Google Scholar 

  269. Gong Q, Wang H, Yu P, Qian T, Xu X. Protective or harmful: the dual roles of autophagy in diabetic retinopathy. Front Med. 2021;8.

  270. Becker K, Klein H, Simon E, Viollet C, Haslinger C, Leparc G, et al. In-depth transcriptomic analysis of human retina reveals molecular mechanisms underlying diabetic retinopathy. Sci Rep. 2021;11(1):10494.

    CAS  PubMed Central  Google Scholar 

  271. de Faria JML, Dátilo MN. Deficient autophagy contributes to the development of diabetic retinopathy. The Eye and Foot in Diabetes: IntechOpen; 2019.

  272. Intartaglia D, Giamundo G, Conte I. Autophagy in the retinal pigment epithelium: a new vision and future challenges. FEBS J. 2021;289:7199.

    PubMed Central  Google Scholar 

  273. Taki K, Horie T, Kida T, Mimura M, Ikeda T, Oku H. Impairment of autophagy causes superoxide formation and caspase activation in 661 W cells, a cell line for cone photoreceptors, under hyperglycemic conditions. Int J Mol Sci. 2020;21(12):4240.

    CAS  PubMed Central  Google Scholar 

  274. Huang C, Lu H, Xu J, Yu H, Wang X, Zhang X. Protective roles of autophagy in retinal pigment epithelium under high glucose condition via regulating PINK1/Parkin pathway and BNIP3L. Biol Res. 2018;51(1):1–9.

    CAS  Google Scholar 

  275. Zhang X, Li T, Cheng H-J, Wang H, Ferrario CM, Groban L, et al. Chronic GPR30 agonist therapy causes restoration of normal cardiac functional performance in a male mouse model of progressive heart failure: insights into cellular mechanisms. Life Sci. 2021;285: 119955.

    CAS  PubMed Central  Google Scholar 

  276. Wang F, He Q, Gao Z, Redington AN. Atg5 knockdown induces age-dependent cardiomyopathy which can be rescued by repeated remote ischemic conditioning. Basic Res Cardiol. 2021;116(1):1–16.

    CAS  Google Scholar 

  277. Kanamori H, Takemura G, Goto K, Tsujimoto A, Mikami A, Ogino A, et al. Autophagic adaptations in diabetic cardiomyopathy differ between type 1 and type 2 diabetes. Autophagy. 2015;11(7):1146–60.

    CAS  PubMed Central  Google Scholar 

  278. Che Y, Wang ZP, Yuan Y, Zhang N, Jin YG, Wan CX, et al. Role of autophagy in a model of obesity: a long-term high fat diet induces cardiac dysfunction. Mol Med Rep. 2018;18(3):3251–61.

    CAS  PubMed Central  Google Scholar 

  279. Kobayashi S, Liang Q. Autophagy and mitophagy in diabetic cardiomyopathy. Biochimica et Biophysica Acta (BBA) Mol Basis Dis. 2015;1852(2):252–61.

    CAS  Google Scholar 

  280. Clément M, Chappell J, Raffort J, Lareyre F, Vandestienne M, Taylor AL, et al. Vascular smooth muscle cell plasticity and autophagy in dissecting aortic aneurysms. Arterioscler Thromb Vasc Biol. 2019;39(6):1149–59.

    PubMed Central  Google Scholar 

  281. Madrigal-Matute J, de Bruijn J, van Kuijk K, Riascos-Bernal DF, Diaz A, Tasset I, et al. Protective role of chaperone-mediated autophagy against atherosclerosis. Proc Natl Acad Sci. 2022;119(14): e2121133119.

    CAS  PubMed Central  Google Scholar 

  282. Kumariya S, Ubba V, Jha RK, Gayen JR. Autophagy in ovary and polycystic ovary syndrome: role, dispute and future perspective. Autophagy. 2021;17(10):2706–33.

    CAS  PubMed Central  Google Scholar 

  283. Yamamoto A, Mizushima N, Tsukamoto S. Fertilization-induced autophagy in mouse embryos is independent of mTORC1. Biol Reprod. 2014;91(1):7.

    Google Scholar 

  284. Sumarac-Dumanovic M, Apostolovic M, Janjetovic K, Jeremic D, Popadic D, Ljubic A, et al. Downregulation of autophagy gene expression in endometria from women with polycystic ovary syndrome. Mol Cell Endocrinol. 2017;440:116–24.

    CAS  Google Scholar 

  285. Zhang C, Hu J, Wang W, Sun Y, Sun K. HMGB1-induced aberrant autophagy contributes to insulin resistance in granulosa cells in PCOS. FASEB J. 2020;34(7):9563–74.

    CAS  Google Scholar 

  286. Mu Y, Yan W-J, Yin T-L, Zhang Y, Li J, Yang J. Diet-induced obesity impairs spermatogenesis: a potential role for autophagy. Sci Rep. 2017;7(1):1–13.

    Google Scholar 

  287. Cunard R. Endoplasmic reticulum stress in the diabetic kidney, the good, the bad and the ugly. J Clin Med. 2015;4(4):715–40.

    CAS  PubMed Central  Google Scholar 

  288. Xie Z, Lau K, Eby B, Lozano P, He C, Pennington B, et al. Improvement of cardiac functions by chronic metformin treatment is associated with enhanced cardiac autophagy in diabetic OVE26 mice. Diabetes. 2011;60(6):1770–8.

    CAS  PubMed Central  Google Scholar 

  289. Zhou H, Chen Y, Huang S-w, Hu P-f, Tang L-j. Regulation of autophagy by tea polyphenols in diabetic cardiomyopathy. J Zhejiang Univ-Sci B. 2018;19(5):333–41.

    CAS  PubMed Central  Google Scholar 

  290. Akbari H, Hosseini-Bensenjan M, Salahi S, Moazzen F, Aria H, Manafi A, et al. Apelin and its ratio to lipid factors are associated with cardiovascular diseases: a systematic review and meta-analysis. PLoS ONE. 2022;17(8): e0271899.

    CAS  PubMed Central  Google Scholar 

  291. Morissette G, Lodge R, Bouthillier J, Marceau F. Receptor-independent, vacuolar ATPase-mediated cellular uptake of histamine receptor-1 ligands: possible origin of pharmacological distortions and side effects. Toxicol Appl Pharmacol. 2008;229(3):320–31.

    CAS  Google Scholar 

  292. Fallah H, Akbari H, Abolhassani M, Mohammadi A, Gholamhosseinian A. Berberis integerrima ameliorates insulin resistance in high-fructose-fed insulin-resistant rats. Iran J Basic Med Sci. 2017;20(10):1093.

    PubMed Central  Google Scholar 

  293. Abdelaziz DH, Thapa S, Abdulrahman B, Vankuppeveld L, Schatzl HM. Metformin reduces prion infection in neuronal cells by enhancing autophagy. Biochem Biophys Res Commun. 2020;523(2):423–8.

    CAS  Google Scholar 

  294. Mardones P, Rubinsztein DC, Hetz C. Mystery solved: trehalose kickstarts autophagy by blocking glucose transport. Sci Signal. 2016;9(416):fs2.

    Google Scholar 

  295. Wang Q, Ren J. mTOR-Independent autophagy inducer trehalose rescues against insulin resistance-induced myocardial contractile anomalies: role of p38 MAPK and Foxo1. Pharmacol Res. 2016;111:357–73.

    CAS  PubMed Central  Google Scholar 

  296. Paech F, Bouitbir J, Krähenbühl S. Hepatocellular toxicity associated with tyrosine kinase inhibitors: mitochondrial damage and inhibition of glycolysis. Front Pharmacol. 2017;8:367.

    PubMed Central  Google Scholar 

  297. Sheng Z, Ma L, Sun JE, Zhu LJ, Green MR. BCR-ABL suppresses autophagy through ATF5-mediated regulation of mTOR transcription. Blood J Am Soc Hematol. 2011;118(10):2840–8.

    CAS  Google Scholar 

  298. Choi S-S, Kim E-S, Jung J-E, Marciano DP, Jo A, Koo JY, et al. PPARγ antagonist Gleevec improves insulin sensitivity and promotes the browning of white adipose tissue. Diabetes. 2016;65(4):829–39.

    CAS  PubMed Central  Google Scholar 

  299. Kwak HJ, Choi H-E, Jang J, Park SK, Bae Y-A, Cheon HG. Bortezomib attenuates palmitic acid-induced ER stress, inflammation and insulin resistance in myotubes via AMPK dependent mechanism. Cell Signal. 2016;28(8):788–97.

    CAS  Google Scholar 

  300. Ashrafizadeh M, Yaribeygi H, Atkin SL, Sahebkar A. Effects of newly introduced antidiabetic drugs on autophagy. Diabetes Metab Syndr. 2019;13(4):2445–9.

    Google Scholar 

  301. Zhou Y, Guo Z, Yan W, Wang W. Cardiovascular effects of sitagliptin–an anti-diabetes medicine. Clin Exp Pharmacol Physiol. 2018;45(7):628–35.

    CAS  Google Scholar 

  302. Arab HH, Gad AM, Reda E, Yahia R, Eid AH. Activation of autophagy by sitagliptin attenuates cadmium-induced testicular impairment in rats: targeting AMPK/mTOR and Nrf2/HO-1 pathways. Life Sci. 2021;269: 119031.

    CAS  Google Scholar 

  303. Lin Z, Wang Z, Zhou X, Zhang M, Gao D, Zhang L, et al. Discovery of new fluorescent thiazole–pyrazoline derivatives as autophagy inducers by inhibiting mTOR activity in A549 human lung cancer cells. Cell Death Dis. 2020;11(7):1–12.

    CAS  Google Scholar 

  304. Yin J, Gu L, Wang Y, Fan N, Ma Y, Peng Y. Rapamycin improves palmitate-induced ER stress/NF κ B pathways associated with stimulating autophagy in adipocytes. Mediat Inflam. 2015;2015.

  305. Armour SM, Baur JA, Hsieh SN, Land-Bracha A, Thomas SM, Sinclair DA. Inhibition of mammalian S6 kinase by resveratrol suppresses autophagy. Aging (Albany NY). 2009;1(6):515.

    CAS  Google Scholar 

  306. Zhu X, Liu Q, Wang M, Liang M, Yang X, Xu X, et al. Activation of Sirt1 by resveratrol inhibits TNF-α induced inflammation in fibroblasts. PLoS ONE. 2011;6(11): e27081.

    CAS  PubMed Central  Google Scholar 

  307. Kim SH, Kim G, Han DH, Lee M, Kim I, Kim B, et al. Ezetimibe ameliorates steatohepatitis via AMP activated protein kinase-TFEB-mediated activation of autophagy and NLRP3 inflammasome inhibition. Autophagy. 2017;13(10):1767–81.

    PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was granted by Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran (Grant No. 401000506).

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

All listed authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship of this article, take responsibility for the integrity of the work as a whole, and have given their approval for this version to be published.

Corresponding author

Correspondence to Hamed Akbari.

Ethics declarations

Ethics approval and consent to participate

This article is based on previously conducted studies and none of the authors have performed any studies on human participants or animals and it is approved by the Ethics Committee of Kerman University of Medical Sciences, Kerman, Iran (IR.KMU.REC.1401.449).

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sadeghi, A., Niknam, M., Momeni-Moghaddam, M.A. et al. Crosstalk between autophagy and insulin resistance: evidence from different tissues. Eur J Med Res 28, 456 (2023). https://doi.org/10.1186/s40001-023-01424-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-023-01424-9

Keywords