Next Article in Journal
Nickel-Atom Doping as a Potential Means to Enhance the Photoluminescence Performance of Carbon Dots
Next Article in Special Issue
Composition of Polygonatum zanlanscianense Pamp. Steam and Leaf Phenolic Extract and Its Protective Mechanism on t-BHP-Induced Oxidative Damage of HepG2 Cells
Previous Article in Journal
Application of Chromatographic and Spectroscopic-Based Methods for Analysis of Omega-3 (ω-3 FAs) and Omega-6 (ω-6 FAs) Fatty Acids in Marine Natural Products
Previous Article in Special Issue
Neuroprotective and Antiherpetic Properties of Polyphenolic Compounds from Maackia amurensis Heartwood
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Drugs: A New Direction for the Prevention and Treatment of Diabetes

Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250001, China
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(14), 5525; https://doi.org/10.3390/molecules28145525
Submission received: 11 May 2023 / Revised: 11 July 2023 / Accepted: 14 July 2023 / Published: 20 July 2023
(This article belongs to the Special Issue Antioxidant Activity of Plant Phenolics)

Abstract

:
Insulin resistance, as a common pathological process of many metabolic diseases, including diabetes and obesity, has attracted much attention due to its relevant influencing factors. To date, studies have mainly focused on the shared mechanisms between mitochondrial stress and insulin resistance, and they are now being pursued as a very attractive therapeutic target due to their extensive involvement in many human clinical settings. In view of the complex pathogenesis of diabetes, natural drugs have become new players in diabetes prevention and treatment because of their wide targets and few side effects. In particular, plant phenolics have received attention because of their close relationship with oxidative stress. In this review, we briefly review the mechanisms by which mitochondrial stress leads to insulin resistance. Moreover, we list some cytokines and genes that have recently been found to play roles in mitochondrial stress and insulin resistance. Furthermore, we describe several natural drugs that are currently widely used and give a brief overview of their therapeutic mechanisms. Finally, we suggest possible ideas for future research related to the unique role that natural drugs play in the treatment of insulin resistance through the above targets.

Graphical Abstract

1. Introduction

Diabetes mellitus is associated with metabolic disorders, with 25% of patients progressing to several microvascular and macrovascular complications, followed by blindness, renal failure, myocardial infarction, or stroke within 20–40 years [1]. Diabetes mellitus can also progress to diabetic ketoacidosis in 10% of cases, which is the leading cause of diabetes mellitus-related death worldwide. As type 2 diabetes (T2DM) is a major burden on healthcare systems, its mechanism has been widely studied for many years, and the most important mechanism leading to T2DM has been shown to be insulin resistance [2]. The term insulin resistance refers to the decreased efficiency of insulin in promoting glucose uptake and utilization in tissues involved in glucose homeostasis for various reasons, for which the body compensates by secreting too much insulin to produce hyperinsulinemia to maintain the stability of blood glucose. Patients with insulin resistance often have other abnormal states, such as impaired glucose tolerance (IGT) and impaired fasting glucose. Glucose tolerance depends on the insulin secretion of the pancreas on the one hand and on the target organ’s sensitivity to insulin, both of which determine whether insulin can work at the right time. When the body develops impaired glucose tolerance, the risk of developing diabetes is greatly increased [3]. Some clinical studies have shown that muscle insulin resistance is more obvious than liver insulin resistance in IGT subjects, and insulin secretion is significantly impaired during the whole period of diabetes [4]. Decades of extensive research results and clinical trials have provided a detailed protocol for the treatment of these diseases, but the specific mechanism remains unclear, greatly limiting early intervention and the prevention and treatment of complications.
A growing recognition of the integrality of metabolic physiology has led to research into the mechanisms and related factors influencing insulin resistance. As early as 1936, Himsworth clarified the concept of insulin resistance. Initially, it was assumed that reduced insulin receptor binding was responsible for the typical obesity-related insulin resistance, but this hypothesis was soon replaced by a model centered on insulin signal transduction defects. The mechanism of insulin resistance is complex, and its pathological changes can also lead to a series of diseases. Most of these diseases are caused by a combination of insulin resistance in skeletal muscle, liver, and fat. Insulin resistance [5] significantly impairs insulin-induced glycogen synthesis in the liver and glucose uptake in muscle and fat. In addition, adipose insulin resistance can lead to excessive triglyceride levels in the liver of patients with non-alcoholic fatty liver disease by promoting the reesterification of circulating fatty acids.
Various research groups have developed new drugs based on mechanisms that have been discovered in recent years. For instance, in 2005, Burkey et al. [6] realized that peptidyl peptidase-IV can improve insulin resistance and help in the treatment of diabetes. Regarding the crosstalk between insulin-responsive tissues, which is one of the complex mechanisms associated with insulin resistance in vivo, Klymenko et al. [7] have carried out relevant expositions and experiments over the years. Traditional thiazolidinediones (TZDs) or metformin, sodium-dependent glucose transporter 2 (SGLT-2) inhibitors, and other medications can have undesirable side effects [8,9,10], while natural drugs have shown unique advantages due to their wide range of targets, few side effects and mild efficacy. Many studies have shown that natural drugs such as cassia semen, rhubarb, aloe vera, and senna can improve insulin resistance [11,12,13].
As early as 2016, Jeong, E. M.’s group [14] demonstrated the beneficial effects of ameliorating mitochondrial oxidative stress on insulin resistance and diastolic dysfunction by treatment with the mitochondria-targeting antioxidant MitoTEMPO. Liver, muscle, and fat are the three major organs associated with peripheral insulin resistance, among which muscle and fat insulin resistance have been pointed out to be closely related to mitochondrial dysfunction [15,16]. The liver is the starting point of insulin resistance and plays a central regulatory role in glucose and lipid metabolism [17]. Moreover, the liver is rich in mitochondria, which, as the “energy factory” of cells, are the main site where reactive oxygen species (ROS) are produced. When ROS are produced faster than they can be removed by the body [18], mitochondrial stress and energy metabolism disorders follow, and they play an important role in the early onset of a variety of metabolic diseases [19,20]. This makes it possible to further explore the specific mechanisms of hepatic insulin resistance.
Overall, in our review, we will introduce the newly discovered cytokines and genes identified by various research groups that are closely related to mitochondrial stress and insulin resistance. This review will also provide a unique summary of some natural drugs for the treatment of insulin resistance-related targets to pave the way for future research directions (Figure 1).
Natural drugs are an important means of prevention and treatment of insulin resistance-related diseases such as diabetes. Insulin resistance is closely related to many pathological processes, such as abnormal insulin signaling pathway conduction, energy metabolism regulation, and inflammation, and emodin, aloe-emodin, chrysol, and beranin, as the main active components of many natural drugs, play a role by targeting one or more targets in the above pathological processes.

2. Mitochondrial Stress, a Key Inducer of Insulin Resistance

Mitochondria have attracted increasing attention due to their complex functions, unique genome, and important role in energy metabolism [21]. In the physiological state, mitochondria regulate cell apoptosis and maintain body homeostasis through their own replication, translation, fusion and fission, mitophagy, and other important factors [22,23]. Under pathological conditions, mitochondrial dysfunction damages cellular homeostasis and creates conditions for the occurrence of a variety of diseases [24,25]. The prevailing view is that mitochondrial defects can be caused by mutations in the mitochondrial genome or by chronic exposure to proinflammatory cytokines, including type I interferon [26]. Mutations in the mitochondrial genome often lead to the appearance of defective proteins and mis-targeted proteins in the mitochondrial proteome, thereby mediating the inactivation or overactivation of the proteasome. In an abnormal state, the damaged homeostasis of the body will first seek to correct itself. For example, mitophagy, which develops from mitochondrial fission, is a way for the body to remove abnormal mitochondria [27]. When mitochondrial dysfunction exceeds the compensatory limit of the body, mitochondrial stress follows, which is manifested as a decrease in the mitochondrial inner membrane electrochemical (IM) potential mainly through the activity of the respiratory chain and the accumulation of ROS caused by dysfunction of the respiratory chain [28]. Mitochondrial self-repair and programmed death are not two independent processes. In the study by Wu, B. et al. [29], it was observed that energy deficiency caused by decreased mitochondrial ATP synthesis stimulates AMP-activated protein kinase (AMPK), which partially restores mitochondrial energy metabolism and biogenesis but also induces mitophagy. Overall, the origin of mitochondrial defects is complex, and if uncorrected, these defects can progress to the next stage of mitochondrial stress.
As mentioned earlier, the pathological process of insulin resistance in the liver is closely related to mitochondrial stress. Mitochondrial stress and insulin resistance form a vicious cycle that jointly promotes the development of the disease. In recent years, people have gradually turned their attention toward the mechanism of mitochondrial stress, hoping to treat metabolic diseases with this as a target. In Figure 2, we visualize some of the factors that play an important role in mitochondrial stress and insulin resistance and point out some possible bridging factors between the two.

2.1. Major Factors Leading to Mitochondrial Stress

Many research groups worldwide have discussed the mechanisms involved in mitochondrial stress. ROS, the mediators of mitochondrial stress, have long been thought to play an important role in neurodegenerative diseases and cardiovascular diseases. As early as 1994, Paolisso et al. [30] uncovered a relationship between mitochondrial stress and metabolic diseases by measuring O2 levels in the bodies of non-insulin-dependent diabetic patients. Over the next two decades, various research teams have proposed several related factors that affect mitochondrial stress. Among them, the most representative and most likely therapeutic targets include PPARs, CypD, and AGEs, which we will discuss in the following sections.
PPARs, activated receptor ligands of the nuclear hormone receptor family that are classified into three subtypes and have been found to control many intracellular metabolic processes, are ligand-inducible nuclear receptors. PPAR-β/δ inhibits mitochondrial stress and reduces ROS production on the one hand and reduces inflammation and insulin resistance induced by interleukin-6 on the other hand, thereby delaying the disease progression of obesity, T2DM, NASH, and other diseases [31,32]. In 2015, Lee et al. [33] noted that PPAR-β/δ of the PPAR family has remarkable effects on improving hepatic insulin sensitivity and reducing insulin resistance and blood sugar levels. Knockout or low expression of PPAR-γ significantly reduced macrophage activation, putting mice at increased risk of obesity and insulin resistance. In summary, PPARs play a role in fatty acid decomposition, cholesterol transport, and energy metabolism, suggesting that PPARs can be an important treatment option for some diseases.
Some studies have mentioned that the key molecule CypD plays an important role in regulating mitochondrial stress. The mitochondrial permeability transition pore (MPTP) is a nonselective channel in the mitochondrial intima. Under physiological conditions, periodic opening and closing are essential for maintaining cell homeostasis [34]. However, an abnormal increase in or modification (acetylation) of the protein expression of the key mitochondrial stress protein CypD leads to excessive irreversible opening of the MPTP [35], causing mitochondrial swelling and dysfunction of the mitochondrial respiratory chain (impaired activity of mitochondrial complex enzymes I–V) and leading to mitochondrial stress and energy metabolism disorders. In 2019, Castillo et al. [36] showed that after CypD caused MPTP opening, the presence of nonesterified fatty acids led to proton leakage from the islets. Changes in CypD expression or acetylation are considered key factors in the regulation of mitochondrial stress; protein acetylation modification, as one of the important aspects of epigenetics, has been a hot research topic in recent years. Recently, we found that in an insulin resistance mouse model induced by a high-fat diet, the luciferase activity and protein expression of CypD in the liver were increased compared with those in the control group [18]. Moreover, CypD gene knockout reduced liver mitochondrial stress, thereby alleviating insulin resistance.
Two molecules that affect mitochondrial stress have thus far been described. Therefore, let us look at the compounds involved. AGEs are stable covalent compounds generated by the spontaneous reaction of macromolecular substances such as proteins, lipids, or nucleic acids with glucose or other reduced monosaccharides without the involvement of enzymes. Numerous research teams have found that AGEs accelerate aging in the human body; that is, they are closely linked to the development of numerous chronic degenerative diseases [37]. The AGE-RAGE axis activates NADPH oxidases, which are the major endogenous sources of ROS, thus aggravating the occurrence of mitochondrial stress [37]. The above process mainly occurs through the mitochondrial respiratory chain and occasionally occurs through stress-related signaling pathways, such as Jun N-terminal kinase and p38. NF-κB activated by AGEs also increases the expression of certain cytokines, such as those that promote inflammation, and the increase in inflammation in the body leads to the exacerbation of disease [17]. AGEs have long been used as biomarkers or predictors of diabetes complications; however, in 2020, Shen et al. [38] showed that the promotion of ROS by AGEs implies that AGEs have a role in the treatment of diabetes, NASH, and other diseases.
In addition to these three factors, other molecules and compounds that cause mitochondria to produce too many ROS are being discovered. Cytochrome P450 family 2 subfamily E member 1 (CYP2E1), located on chromosome 10, is mainly involved in the metabolism of low-molecular-weight substances. High insulin levels in the body and other pathological changes, such as steatosis, can promote the body’s ability to increase the production of ROS [39]. The oxidizing capacity of iron also contributes to the production of ROS [40]. In addition, some recent studies have shown that the activation of NADPH oxidase 4 (NOX4) promotes ROS production while prolonging the endoplasmic reticulum stress mediated by OS, which leads to liver cell apoptosis and accelerates the progression of NASH and other diseases [41]. In addition, many experiments have shown that liver-specific PGC-1 deficiency expands the influence of mitochondrial stress and accelerates the development of NASH in mice, suggesting that PGC-1 inhibits oxidative stress and inflammation, leading to improvement in disease [42]. In terms of compounds, in 2016, Chakraborti et al. [40] showed that a diet rich in fructose greatly speeds up the production of ROS.
These factors usually regulate insulin resistance by affecting mitochondrial stress, but some factors not only mediate or inhibit mitochondrial stress but also aggravate or improve insulin resistance themselves. For example, many scholars have demonstrated that microRNAs can reduce insulin resistance either directly [43] or indirectly (by inhibiting mitochondrial stress and so on) [44]. MicroRNAs, or miRNAs for short, are a class of endogenous small noncoding RNAs with regulatory functions found in eukaryotes. Several articles have described the mechanisms of these small RNAs in various types of diabetes and other metabolic diseases [45,46]. miR-155-5p protects beta cells from insulin resistance by promoting insulin secretion by the body’s beta cells in the presence of glucose [47]. Moreover, miR-126 and miR-30c are two important benign factors in patients with diabetic cardiomyopathy. The former indirectly activates sirtuin-1 (SIRT1) and superoxide dismutase (SOD) to induce resistance to oxidative stress, while the latter targets PGC-1, not only inducing PPAR-α but also playing a positive role in key mitochondrial regulation. Together, the two can reduce excessive ROS and myocardial lipid accumulation in patients with diabetic cardiomyopathy, thereby preventing pathological processes such as cardiac insufficiency [48]. miRNAs have been applied in various fields because of their newly discovered wide use. At present, most in vitro experiments on reducing the effects of insulin resistance and treating diabetes with miRNAs have been successful, but the therapeutic effects of miRNAs in vivo remain to be further studied.

2.2. How Does Mitochondrial Stress Affect Insulin Resistance?

Based on some clinical studies, a variety of drugs have also been observed to act by targeting mitochondrial dysfunction. Empagliflozin (EMPA) [49] and pyridostigmine (PYR) [50] have good preventive effects on diabetic heart damage because they can regulate the shape and function of mitochondrial cristae, thereby improving mitochondrial function in heart tissue, and can regulate mitochondrial fusion and fission. Wang, X. et al. [51] observed that zinc finger, BED-type containing 6 (ZBED6) may target the abnormal accumulation of mitochondrial ROS, thereby improving insulin resistance in the body. The therapeutic effects of some natural medicines on diabetes are also dependent on the regulation of mitochondrial function, including astaxanthin (AX) treatment, which stimulates mitochondrial biogenesis by activating the AMPK pathway in skeletal muscle and significantly improves insulin resistance [52]. Many studies have confirmed that the relationship between glucose tolerance, insulin resistance, and mitochondrial dysfunction may not be direct. Factors that play a role in this process would be effective targets for drugs to prevent and treat diseases related to insulin resistance, which is why investigating how mitochondrial stress leads to insulin resistance has been the focus of many research groups.

2.2.1. HSP60

Heat shock protein 60 (HSP60) is a chaperone protein that is overexpressed when mitochondrial stress occurs. This state not only impairs insulin signaling and insulin sensitivity in mouse liver cells but also promotes endoplasmic reticulum stress, which induces liver adipogenesis and insulin resistance. In 2020, Xiao et al. [53] presented experimental evidence for this claim. In their experiment, HSP60 knockout inhibited the mammalian target of rapamycin complex 1—sterol-regulatory-element-binding protein 1 (mTORC1-SREBP1) signaling and hepatic adipogenesis, suggesting that HSP60 may be involved in endoplasmic reticulum stress-induced mTORC1-SREBP1 signal transduction related to adipogenesis and steatosis. Therefore, the researchers concluded that mitochondrial stress and endoplasmic reticulum stress coregulate the insulin resistance process in the body.
However, HSP60 acts differently in different parts of the body. On the one hand, as described previously, miRNAs play an important role in influencing insulin resistance. On the other hand, a high serum level of HSP60 is often accompanied by the downregulation of HSP60 in the myocardium, which means that the inhibition of HSP60 in the myocardium will lead to the intensification of insulin resistance [54]. In 2020, Wen et al. [38] found that miR-802-5p causes cardiac insulin resistance by reducing HSP60 expression. HSP60 is also an important therapeutic target for insulin resistance in the heart, as researchers have recently discovered.

2.2.2. FGF21

Does mitochondrial stress under pathological conditions necessarily result in insulin resistance? In 2013, Kim argued against this. Autophagy-related 7 (ATG7) is a gene that is critical for autophagosome activity; in the ATG7 knockout mouse, Kim et al. [55] were surprised to observe that the mice were not fat but lean and were unaffected by obesity and insulin resistance induced by a high-fat diet. Inhibition of autophagy leads to mitochondrial stress; however, this mitochondrial stress improves insulin resistance. They further explored the bridging factor between the two. In a series of experiments, they identified a factor called fibroblast growth factor 21 (FGF21), which once served as a marker for a variety of mitochondrial diseases caused by mutations in mitochondrial DNA (mtDNA) [56,57]. Researchers have found that inhibiting the expression of mitochondrial oxidative phosphorylation genes results in damage to the mitochondrial respiratory chain and a reduction in ATP, thereby activating the integrated stress response (ISR) and mediating the production of FGF21 [58,59]. Similar to the HSP60 knockout results, FGF21 also improves hepatic insulin sensitivity by inhibiting mTORC1. Moreover, FGF21 activation increases fatty acid oxidation, lipolysis, and the browning of white adipose tissue in mice, which protects mice from obesity and insulin resistance. The researchers also observed that metformin administration increases serum levels of FGF21, suggesting that FGF21 is induced by metformin, providing indirect evidence of FGF21’s therapeutic value [60].
Currently, FGF21 and its analogs have been used in laboratory animals and human subjects to significantly improve lipid distribution in the body and to reduce the body weight of subjects, which is why it has become a promising new drug for the treatment of obesity, diabetes, and metabolic syndrome. Unfortunately, in addition to the mitochondrial stress mentioned above, FGF21 can also be induced by a variety of stresses, such as obesity, exercise or cold exposure, and the PPAR family. The connection between these obvious nonmitochondrial inducers of FGF21 and mitochondrial stress is still unclear and needs to be uncovered.

2.2.3. GDF-15

Stress response protein growth differentiation factor-15 (GDF-15) is secreted during mitochondrial stress or functional damage [61,62,63], and high levels of GDF-15 are closely associated with a variety of pathological diseases, including inflammation, cancer, and various mitochondrial diseases [64], which is why GDF-15 has been extensively explored as a biomarker of mitochondrial disease and is thought to be closely related to the mitochondrial function of patients [65,66,67]. The increase in GDF-15 in the body has been proven to be induced by dysfunction of the mitochondrial respiratory chain rather than other metabolic dysfunctions [68]. Furthermore, Straub [69] suggested a potential connection between mitochondrial respiratory dysfunction, the NADH/NAD ratio, impaired ATP levels, and GDF-15. Steffen, J. et al. [70] showed that knockout of drp leading to ER stress and mitochondrial abnormalities can activate the AFT4-controlled integrated stress response (ISR) and increase the hepatic expression of GDF-15, which subsequently aggravates inflammation, fibrosis, and necrosis. This result implies that GDF-15 regulates its own expression in response to impaired mitochondrial function through the ISR pathway, just as the ISR pathway also induces GDF-15 hyperexpression during metformin treatment. Treatment with GDF-15 and metformin induces a process that is also related to multiple pathways. Aguilar-Recarte et al. [71] showed that metformin can maintain full AMPK activation by upregulating GDF-15 to participate in the therapeutic process of this drug. The study by Zhang, S. Y. [72] also observed that metformin regulates energy homeostasis through the renal GDF-15-dependent AP axis. In conclusion, the above results indicate that mitochondrial stress caused by mitochondrial dysfunction induces an increase in GDF-15. GDF-15 can not only be used as a marker of some metabolic diseases but can also play an important role in the diagnosis and treatment of diseases such as diabetes mellitus.
The relationship between GDF-15 and insulin resistance has also been explored by some research groups. Several studies in recent years have found that GDF-15 can be used as a therapeutic target for obesity and prediabetic glucose tolerance disorders. Studies have shown that GDF-15 is associated with obesity and insulin resistance, abnormal blood glucose levels, and impaired blood glucose homeostasis in the body. In 2017, Chung et al. [73] used recombinant GDF-15 in ob/ob mice and found that the mice lost weight and exhibited improved insulin sensitivity. In 2020, Choi [62] found that GDF-15 and FGF21 secretion was significantly upregulated in adipocyte-specific Crif1 (also known as Gadd45gip1) knockout mice on a high-fat diet, which was able to inhibit weight gain and improve glucose tolerance. Recently, the relationship and the specific mechanism between GDF-15 and insulin resistance have received increasing attention.
GDF-15 shows complex effects in different stages of disease; however, its specific mechanism in the early onset of metabolic diseases, especially hepatic insulin resistance, is still unclear. As we mentioned earlier, CypD plays an important role in the regulation of mitochondrial stress, and there is a large body of evidence to suggest this; at the same time, our group’s previous study suggested that GDF-15 is downstream of CypD. Therefore, in this review, we propose a potential mechanism that describes the possible relationship between CypD and GDF-15. Increased expression or activity of CypD causes mitochondrial stress and energy metabolism disorders, which may affect insulin resistance by regulating the stress response protein GDF-15. The specific mechanism remains to be further studied (Figure 3).

3. Complex Mechanisms of Natural Drugs in the Treatment of Diseases Related to Insulin Resistance

Plant-derived secondary metabolites [74] are organic compounds produced by plants and are commonly known as a class of natural drugs. Plant-derived secondary metabolites are more readily available and safer than conventional chemical drugs [74] and have been shown to have clinically meaningful and mild efficacy in cancer treatment, inflammation reduction, and diabetes treatment. To date, researchers have identified more than 400,000 secondary metabolites in nature [75], including allicin, quercetin, eugenol, lycorine, tea polyphenols, and berberine, and more new plant-derived active compounds are being discovered. Among them, these active compounds can be widely classified into terpenoids and their derivatives, alkaloids, steroids, amino acids, polysaccharide antimicrobial peptides, lignans, saponins, and new structures [75]. Recently, an increasing number of research teams have begun to pay attention to the effect of secondary metabolites on insulin resistance and related mechanisms in the pathogenesis of diabetes.
Many studies have confirmed the good hypoglycemic ability of a variety of plant derivatives. Moreover, numerous clinical cases have verified the therapeutic effect of resveratrol on diabetic patients [76]. Fasting blood glucose and serum insulin levels were significantly reduced during the treatment, and insulin sensitivity and glucose and lipid metabolism of the body were also restored [77]. Similar to the way that resveratrol exerts its therapeutic effect, curcumin, as a new drug for obesity and diabetes, can also alleviate the progression of insulin resistance through AMPK, nuclear factor erythroid 2-related factor 2 (Nrf2) and PPAR-related molecular pathways [78,79], which may involve endoplasmic reticulum stress and other oxidative stress abnormalities [80]. This process is closely related to the regulation of the intestinal flora [81]. However, anthocyanins (anthocyanin 3-glucoside) and proanthocyanidins (PCs), which are widely present in black bean seeds, have been shown in animal experiments to activate AMPK- and glucose transporter type 4 (GLUT4)-related pathways, alleviate hyperglycemia, and restore insulin sensitivity in mice [82]. In addition to the AMPK-related pathway, targeting other important factors in the regulation of insulin resistance can achieve similar therapeutic effects. The abnormal activation of IRS often leads to impairment in the insulin signaling pathway and then mediates the occurrence of insulin resistance through inflammatory factors such as tumor necrosis factor (TNF)-α [83]. Carnosic acid and rosmarinic acid can reduce the occurrence of insulin resistance by regulating the AMPK pathway and IRS activation [84,85,86]. In addition, Carica papaya extracts are equally rich in plant polyphenols. It has been confirmed that they reduce blood glucose and lipid levels, and the mechanism may be related to the improvement in endothelial NO synthase (eNOS), which is related to the accumulation of ROS and energy imbalance [87]. In summary, plant polyphenols are beneficial in the treatment of insulin resistance, and the mechanism is often closely related to abnormal insulin signaling mediated by oxidative stress.
Anthraquinone natural products, including emodin, aloe-emodin, rhein, chrysophanol, aurantio-obtusin, and alaternin, are the main active ingredients of many natural drugs. Existing studies have found that they can alleviate insulin resistance from many aspects, such as regulating the insulin signaling pathway, regulating energy homeostasis, and improving inflammation. Emodin has been shown to increase the phosphorylation of AMPK protein and acetyl-CoA carboxylase (ACC) protein in the liver, upregulate the expression of carnitine palmitoyl transferase 1 (CPT1), and downregulate the expression of SREBP-1c and fatty acid synthase (FAS), thereby improving energy metabolism disorders, reducing fat accumulation, and promoting cellular glucose absorption. Emodin can reduce the levels of fasting blood glucose and fasting insulin in mice and can improve insulin resistance [88,89]. Moreover, emodin can promote M2 polarization of macrophages by increasing triggering receptor expressed on myeloid cells 2 (TREM2) expression, significantly reduce local and systemic inflammatory responses in obese mice, inhibit weight gain and lipid accumulation, and reduce fasting blood glucose and fasting insulin levels [90]. Aloe-emodin has also been found to be effective in reducing the production of the inflammatory factors TNF-α and interleukin (IL)-6 and in suppressing the NF-κB signaling pathway, thereby restoring insulin signaling, lowering fasting blood glucose, regulating islet β-cell function, and inhibiting fat accumulation to reduce obesity and insulin resistance. Aloe-emodin has been shown to alleviate inflammation [13,91,92,93]. Rhein can also improve insulin resistance by reducing adipose tissue inflammation and liver triglyceride accumulation in mice [94]. In addition, rhein promotes macrophage polarization toward the M2 phenotype [95], which can reduce inflammation and thus alleviate insulin resistance [96]. Cassia seed, a natural drug rich in anthraquinone products such as aurantio-obtusin and alaternin, can reduce the fasting blood glucose and fasting insulin contents in obese mice and restore insulin sensitivity by increasing skeletal muscle glucose uptake in obese mice [97]. The targets of aurantio-obtusin are more diverse. On the one hand, it phosphorylates AMPK, an important pathway for energy homeostasis. On the other hand, the expression of FAS is inhibited to reduce fat synthesis and thus improve insulin resistance [98]. In addition, aurantio-obtusin can activate the phosphoinositide 3-kinase (PI3K)–protein kinase B (AKT) signaling pathway in liver and adipose tissue, reduce fasting blood glucose, and improve glucose tolerance [99]. Alaternin also plays a role in regulating insulin signaling. Alaternin regulates downstream signals of insulin and restores insulin signaling by competitive inhibition of protein tyrosine phosphatase 1B (PTP1B) [100].
Among natural drugs, other natural products besides plant polyphenols and anthraquinones have shown promising results in the treatment of insulin resistance. Allicin, an organic sulfur compound extracted from the bulb of Allium garlic, has been shown to be effective in the treatment of various diseases due to its specific actions, including antibacterial, antioxidative stress, regulation of cardiovascular and cerebrovascular diseases, reduction in blood lipids, and regulation of diabetes. One study showed that allicin had a hypoglycemic effect in type 2 diabetes patients and animal models in which it could activate pancreatic insulin secretion and reduce total cholesterol (TC), triglyceride (TG), and fasting blood glucose (FBG) levels [101]. Zhai et al. [102] showed that the hypoglycemic activity of allicin in diabetic rats was similar to that of glibenclamide and insulin. In addition, similar to the way in which allicin acts, allyl propyl disulfide, cysteine sulfoxide, and S-allyl cysteine sulfoxide prevent liver-induced insulin activation, increase the insulin production capacity of islet β-cells, separate insulin from its bound form, and increase the insulin sensitivity of cells. The subsequent effect is to reduce blood glucose levels [103,104]. Therefore, the use of bioactive compounds derived from natural sources has become a new direction for the treatment of diabetes due to the moderate and obvious therapeutic effects of these metabolites, which is very consistent with the idea of treating chronic diseases.
The side effects caused by drugs in the process of disease prevention have always been the focus of academic attention, which is why early intervention for diabetes is basically based on exercise for prevention and dietary intervention. Based on the low side effects and high safety characteristics of natural medicines, we speculate that natural medicines play a key role in disease prevention and are expected to be used as an adjunct to prevention methods such as exercise [105,106]. In addition, the treatment of patients with gestational diabetes mellitus requires that close attention be given to the effects of drugs on the mother and fetus, as natural drugs may also have unexpected effects. We further hypothesized that oral natural medicines might improve glucose metabolism through the regulation of the gut microbiota, and a recent study preliminarily confirmed our hypothesis [107]. We also found that Leyva-Gomez, G. et al. [108] proposed the use of natural medicine-rich films to promote wound healing, which may be a new application for the prevention and treatment of diabetic foot. Although the extraction of some natural products is limited by technology and cost [109], we believe that natural products will become new stars in various fields.

4. Conclusions and Future Perspectives

In this article, we introduced the mechanisms of mitochondrial stress and insulin resistance and the high-profile inducing or inhibiting factors, and importantly, we proposed that CypD may lead to abnormal mitochondrial stress and affect insulin resistance through GDF-15, which represents a direction for future research (Table 1).
Currently, an increasing number of drugs have been applied to the treatment of diseases related to glucose and lipid metabolism disorders. Throughout years of exploring the efficacy and safety of various diabetes drugs, natural drugs have shown unique advantages. We thus put forward a good prospect: In the future, natural drugs will become the new favored approach in the process of diabetes prevention and treatment due to their high safety profile and broad action targets. Plant polyphenols, in particular, are closely related to oxidative stress and alleviate the progression of diabetes caused by insulin resistance through the regulation of the insulin signaling pathway. We look forward to more research teams discovering additional natural drugs, and we hope to further explore the specific mechanisms by which natural drugs work, which represents a research direction with great potential.

Author Contributions

Writing—Original draft preparation, P.W.; Writing—Review & editing, P.W. and X.W.; Supervision, X.W.; Funding acquisition, X.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the China National Natural Science Foundation, grant number 81900736; China Postdoctoral Science Foundation, grant number 2023M732137; Shandong Province “Double-Hundred Talent Plan” on 100 Foreign Experts and 100 Foreign Expert Teams Introduction, grant number WST2018004 and the Innovation Project of Shandong Academy of Medical Sciences, grant number 2019-11.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

I would like to thank Junjun Liu for his guidance.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

References

  1. Zheng, Y.; Ley, S.H.; Hu, F.B. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat. Rev. Endocrinol. 2018, 14, 88–98. [Google Scholar] [CrossRef] [PubMed]
  2. Kumar, A.; Sundaram, K.; Mu, J.; Dryden, G.W.; Sriwastva, M.K.; Lei, C.; Zhang, L.; Qiu, X.; Xu, F.; Yan, J.; et al. High-fat diet-induced upregulation of exosomal phosphatidylcholine contributes to insulin resistance. Nat. Commun. 2021, 12, 213–234. [Google Scholar] [CrossRef]
  3. Broughton, D.L.; Taylor, R. Review: Deterioration of Glucose Tolerance with Age: The Role of Insulin Resistance. Age Ageing 1991, 20, 221–225. [Google Scholar] [CrossRef] [PubMed]
  4. Abdul-Ghani, M.A.; Tripathy, D.; DeFronzo, R.A. Contributions of beta-Cell Dysfunction and Insulin Resistance to the Pathogenesis of Impaired Glucose Tolerance and Impaired Fasting Glucose. Diabetes Care 2006, 29, 1130–1139. [Google Scholar] [CrossRef] [PubMed]
  5. Perseghin, G.; Price, T.B.; Petersen, K.F.; Roden, M.; Cline, G.W.; Gerow, K.; Rothman, D.L.; Shulman, G.I. Increased Glucose Transport–Phosphorylation and Muscle Glycogen Synthesis after Exercise Training in Insulin-Resistant Subjects. N. Engl. J. Med. 1996, 335, 1357–1362. [Google Scholar] [CrossRef]
  6. Burkey, B.F.; Li, X.; Bolognese, L.; Balkan, B.; Mone, M.; Russell, M.; Hughes, T.E.; Wang, P.R. Acute and Chronic Effects of the Incretin Enhancer Vildagliptin in Insulin-Resistant Rats. J. Pharmacol. Exp. Ther. 2005, 315, 688–695. [Google Scholar] [CrossRef] [Green Version]
  7. Klymenko, O.; Brecklinghaus, T.; Dille, M.; Springer, C.; de Wendt, C.; Altenhofen, D.; Binsch, C.; Knebel, B.; Scheller, J.; Hardt, C.; et al. Histone deacetylase 5 regulates interleukin 6 secretion and insulin action in skeletal muscle. Mol. Metab. 2020, 42, 101062. [Google Scholar] [CrossRef]
  8. Beck, G.R.; Khazai, N.B.; Bouloux, G.F.; Camalier, C.E.; Lin, Y.; Garneys, L.M.; Siqueira, J.; Peng, L.; Pasquel, F.; Umpierrez, D.; et al. The effects of thiazolidinediones on human bone marrow stromal cell differentiation in vitro and in thiazolidinedione-treated patients with type 2 diabetes. Transl. Res. 2012, 161, 145–155. [Google Scholar] [CrossRef] [Green Version]
  9. Bonnet, F.; Scheen, A. Understanding and overcoming metformin gastrointestinal intolerance. Diabetes Obes. Metab. 2017, 19, 473–481. [Google Scholar] [CrossRef]
  10. Taylor, S.I.; Blau, J.E.; Rother, K.I.; Beitelshees, A.L. SGLT2 inhibitors as adjunctive therapy for type 1 diabetes: Balancing benefits and risks. Lancet Diabetes Endocrinol. 2019, 7, 949–958. [Google Scholar] [CrossRef]
  11. Ko, E.; Um, M.Y.; Choi, M.; Han, T.; Kim, I.-H.; Shin, S. Cassia tora Seed Improves Pancreatic Mitochondrial Function Leading to Recovery of Glucose Metabolism. Am. J. Chin. Med. 2020, 48, 615–629. [Google Scholar] [CrossRef]
  12. Cheng, F.-R.; Cui, H.-X.; Fang, J.-L.; Yuan, K.; Guo, Y. Ameliorative Effect and Mechanism of the Purified Anthraquinone-Glycoside Preparation from Rheum palmatum L. on Type 2 Diabetes Mellitus. Molecules 2019, 24, 1454. [Google Scholar] [CrossRef] [Green Version]
  13. Deora, N.; Sunitha, M.; Satyavani, M.; Harishankar, N.; Vijayalakshmi, M.A.; Venkataraman, K.; Venkateshan, V. Alleviation of diabetes mellitus through the restoration of β-cell function and lipid metabolism by Aloe vera (L.) Burm. f. extract in obesogenic WNIN/GR-Ob rats. J. Ethnopharmacol. 2021, 272, 113921. [Google Scholar] [CrossRef]
  14. Jeong, E.M.; Chung, J.; Liu, H.; Go, Y.; Gladstein, S.; Farzaneh-Far, A.; Lewandowski, E.D.; Dudley, S.C., Jr. Role of Mitochondrial Oxidative Stress in Glucose Tolerance, Insulin Resistance, and Cardiac Diastolic Dysfunction. J. Am. Heart Assoc. 2016, 5, e003046. [Google Scholar] [CrossRef] [Green Version]
  15. Schrauwen, P.; Schrauwen-Hinderling, V.; Hoeks, J.; Hesselink, M.K. Mitochondrial dysfunction and lipotoxicity. Biochim. Biophys. Acta BBA-Mol. Cell Biol. Lipids 2010, 1801, 266–271. [Google Scholar] [CrossRef]
  16. Choi, K.-M.; Ryan, K.K.; Yoon, J.C. Adipose Mitochondrial Complex I Deficiency Modulates Inflammation and Glucose Homeostasis in a Sex-Dependent Manner. Endocrinology 2022, 163, bqac018. [Google Scholar] [CrossRef]
  17. Nowotny, K.; Jung, T.; Höhn, A.; Weber, D.; Grune, T.; Nowotny, K.; Jung, T.; Höhn, A.; Weber, D.; Grune, T. Advanced Glycation End Products and Oxidative Stress in Type 2 Diabetes Mellitus. Biomolecules 2015, 5, 194–222. [Google Scholar] [CrossRef] [Green Version]
  18. Wang, X.; Du, H.; Shao, S.; Bo, T.; Yu, C.; Chen, W.; Zhao, L.; Li, Q.; Wang, L.; Liu, X.; et al. Cyclophilin D deficiency attenuates mitochondrial perturbation and ameliorates hepatic steatosis. Hepatology 2018, 68, 62–77. [Google Scholar] [CrossRef]
  19. Nassir, F.; Ibdah, J.A. Role of Mitochondria in Nonalcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2014, 15, 8713–8742. [Google Scholar] [CrossRef] [Green Version]
  20. Galloway, C.A.; Lee, H.; Brookes, P.; Yoon, Y. Decreasing mitochondrial fission alleviates hepatic steatosis in a murine model of nonalcoholic fatty liver disease. Am. J. Physiol.-Gastrointest. Liver Physiol. 2014, 307, G632–G641. [Google Scholar] [CrossRef] [Green Version]
  21. Taanman, J.-W. The mitochondrial genome: Structure, transcription, translation and replication. Biochim. Biophys. Acta BBA-Bioenergies 1999, 1410, 103–123. [Google Scholar] [CrossRef] [Green Version]
  22. Gustafsson, C.M.; Falkenberg, M.; Larsson, N.-G. Maintenance and Expression of Mammalian Mitochondrial DNA. Annu. Rev. Biochem. 2016, 85, 133–160. [Google Scholar] [CrossRef] [PubMed]
  23. Adebayo, M.; Singh, S.; Singh, A.P.; Dasgupta, S. Mitochondrial fusion and fission: The fine-tune balance for cellular homeostasis. FASEB J. 2021, 35, e21620. [Google Scholar] [CrossRef] [PubMed]
  24. Supinski, G.S.; Schroder, E.A.; Callahan, L.A. Mitochondria and Critical Illness. Chest 2020, 157, 310–322. [Google Scholar] [CrossRef]
  25. Chan, D.C. Mitochondrial Dynamics and Its Involvement in Disease. Annu. Rev. Pathol. 2020, 15, 235–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Becker, Y.L.C.; Duvvuri, B.; Fortin, P.R.; Lood, C.; Boilard, E. The role of mitochondria in rheumatic diseases. Nat. Rev. Rheumatol. 2022, 18, 621–640. [Google Scholar] [CrossRef]
  27. Kleele, T.; Rey, T.; Winter, J.; Zaganelli, S.; Mahecic, D.; Lambert, H.P.; Ruberto, F.P.; Nemir, M.; Wai, T.; Pedrazzini, T.; et al. Distinct fission signatures predict mitochondrial degradation or biogenesis. Nature 2021, 593, 435–439. [Google Scholar] [CrossRef]
  28. Cadenas, S. Mitochondrial uncoupling, ROS generation and cardioprotection. Biochim. Biophys. Acta (BBA) Bioenerg. 2018, 1859, 940–950. [Google Scholar] [CrossRef]
  29. Wu, B.; Chen, Y.; Clarke, R.; Akala, E.; Yang, P.; He, B.; Gao, H. AMPK Signaling Regulates Mitophagy and Mitochondrial ATP Production in Human Trophoblast Cell Line BeWo. Front. Biosci. 2022, 27, 118. [Google Scholar] [CrossRef]
  30. Paolisso, G.; D’Amore, A.; Volpe, C.; Balbi, V.; Saccomanno, F.; Galzerano, D.; Giugliano, D.; Varricchio, M.; D’Onofrio, F. Evidence for a relationship between oxidative stress and insulin action in non-insulin-dependent (type II) diabetic patients. Metabolism 1994, 43, 1426–1429. [Google Scholar] [CrossRef]
  31. Silva, A.K.S.; Peixoto, C.A. Role of peroxisome proliferator-activated receptors in non-alcoholic fatty liver disease inflammation. Cell. Mol. Life Sci. 2018, 75, 2951–2961. [Google Scholar] [CrossRef]
  32. Chen, J.; Montagner, A.; Tan, N.S.; Wahli, W. Insights into the Role of PPARβ/δ in NAFLD. Int. J. Mol. Sci. 2018, 19, 1893. [Google Scholar] [CrossRef] [Green Version]
  33. Lee, H.J.; Yeon, J.E.; Ko, E.J.; Yoon, E.L.; Suh, S.J.; Kang, K.; Kim, H.R.; Kang, S.H.; Yoo, Y.J.; Je, J.; et al. Peroxisome proliferator-activated receptor-delta agonist ameliorated inflammasome activation in nonalcoholic fatty liver disease. World J. Gastroenterol. 2015, 21, 12787–12799. [Google Scholar] [CrossRef]
  34. Liu, X.; Du, H.; Chai, Q.; Jia, Q.; Liu, L.; Zhao, M.; Li, J.; Tang, H.; Chen, W.; Zhao, L.; et al. Blocking mitochondrial cyclophilin D ameliorates TSH-impaired defensive barrier of artery. Redox Biol. 2018, 15, 418–434. [Google Scholar] [CrossRef]
  35. Hou, D.; Hu, F.; Mao, Y.; Yan, L.; Zhang, Y.; Zheng, Z.; Wu, A.; Forouzanfar, T.; Pathak, J.L.; Wu, G. Cationic antimicrobial peptide NRC-03 induces oral squamous cell carcinoma cell apoptosis via CypD-mPTP axis-mediated mitochondrial oxidative stress. Redox Biol. 2022, 54, 102355. [Google Scholar] [CrossRef]
  36. Castillo, E.C.; Morales, J.A.; Chapoy-Villanueva, H.; Silva-Platas, C.; Treviño-Saldaña, N.; Guerrero-Beltrán, C.E.; Bernal-Ramírez, J.; Torres-Quintanilla, A.; García, N.; Youker, K.; et al. Mitochondrial Hyperacetylation in the Failing Hearts of Obese Patients Mediated Partly by a Reduction in SIRT3: The Involvement of the Mitochondrial Permeability Transition Pore. Cell. Physiol. Biochem. 2019, 53, 465–479. [Google Scholar] [CrossRef] [Green Version]
  37. Moldogazieva, N.T.; Mokhosoev, I.M.; Mel’nikova, T.I.; Porozov, Y.B.; Terentiev, A.A. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. Oxidative Med. Cell. Longev. 2019, 2019, 3085756. [Google Scholar] [CrossRef] [Green Version]
  38. Shen, C.-Y.; Lu, C.-H.; Wu, C.-H.; Li, K.-J.; Kuo, Y.-M.; Hsieh, S.-C.; Yu, C.-L. The Development of Maillard Reaction, and Advanced Glycation End Product (AGE)-Receptor for AGE (RAGE) Signaling Inhibitors as Novel Therapeutic Strategies for Patients with AGE-Related Diseases. Molecules 2020, 25, 5591. [Google Scholar] [CrossRef]
  39. Selenscig, D.; Ferreira, M.D.R.; Chicco, A.; Lombardo, Y.B. Dietary fish oil ameliorates adipose tissue dysfunction in insulin-resistant rats fed a sucrose-rich diet improving oxidative stress, peroxisome proliferator-activated receptor γ and uncoupling protein 2. Food Funct. 2018, 9, 2496–2507. [Google Scholar] [CrossRef]
  40. Sil, R.; Chakraborti, A.S. Oxidative Inactivation of Liver Mitochondria in High Fructose Diet-Induced Metabolic Syndrome in Rats: Effect of Glycyrrhizin Treatment. Phytother. Res. 2016, 30, 1503–1512. [Google Scholar] [CrossRef]
  41. Rabelo, F.; Stefano, J.T.; Cavaleiro, A.M.; Lima, R.V.C.; de Campos Mazo, D.F.; Carrilho, F.J.; Correa-Giannella, M.L.; Oliveira, C.P. Association between the CYBA and NOX4 genes of NADPH oxidase and its relationship with metabolic syndrome in non-alcoholic fatty liver disease in Brazilian population. Hepatobiliary Pancreat. Dis. Int. 2018, 17, 330–335. [Google Scholar] [CrossRef] [PubMed]
  42. Besse-Patin, A.; Léveillé, M.; Oropeza, D.; Nguyen, B.N.; Prat, A.; Estall, J.L. Estrogen Signals Through Peroxisome Proliferator-Activated Receptor−γ Coactivator 1α to Reduce Oxidative Damage Associated with Diet-Induced Fatty Liver Disease. Gastroenterology 2017, 152, 243–256. [Google Scholar] [CrossRef] [PubMed]
  43. Berry, C.; Lal, M.; Binukumar, B.K. Crosstalk Between the Unfolded Protein Response, MicroRNAs, and Insulin Signaling Pathways: In Search of Biomarkers for the Diagnosis and Treatment of Type 2 Diabetes. Front. Endocrinol. 2018, 9, 210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Qadir, M.M.F.; Klein, D.; Álvarez-Cubela, S.; Domínguez-Bendala, J.; Pastori, R.L. The Role of MicroRNAs in Diabetes-Related Oxidative Stress. Int. J. Mol. Sci. 2019, 20, 5423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Dotta, F.; Ventriglia, G.; Snowhite, I.V.; Pugliese, A. MicroRNAs: Markers of beta-cell stress and autoimmunity. Curr. Opin. Endocrinol. Diabetes 2018, 25, 237–245. [Google Scholar] [CrossRef]
  46. LaPierre, M.P.; Stoffel, M. MicroRNAs as stress regulators in pancreatic beta cells and diabetes. Mol. Metab. 2017, 6, 1010–1023. [Google Scholar] [CrossRef]
  47. Zhu, M.; Wei, Y.; Geißler, C.; Abschlag, K.; Campos, J.C.; Hristov, M.; Möllmann, J.; Lehrke, M.; Karshovska, E.; Schober, A. Hyperlipidemia-Induced MicroRNA-155-5p Improves β-Cell Function by Targeting Mafb. Diabetes 2017, 66, 3072–3084. [Google Scholar] [CrossRef] [Green Version]
  48. Yin, Z.; Zhao, Y.; He, M.; Li, H.; Fan, J.; Nie, X.; Yan, M.; Chen, C.; Wang, D.W. MiR-30c/PGC-1β protects against diabetic cardiomyopathy via PPARα. Cardiovasc. Diabetol. 2019, 18, 7. [Google Scholar] [CrossRef]
  49. Wang, J.; Huang, X.; Liu, H.; Chen, Y.; Li, P.; Liu, L.; Li, J.; Ren, Y.; Huang, J.; Xiong, E.; et al. Empagliflozin Ameliorates Diabetic Cardiomyopathy via Attenuating Oxidative Stress and Improving Mitochondrial Function. Oxidative Med. Cell. Longev. 2022, 2022, 1122494. [Google Scholar] [CrossRef]
  50. Xue, R.-Q.; Yu, X.-J.; Zhao, M.; Xu, M.; Wu, Q.; Cui, Y.-L.; Yang, S.; Li, D.-L.; Zang, W.-J. Pyridostigmine alleviates cardiac dysfunction via improving mitochondrial cristae shape in a mouse model of metabolic syndrome. Free. Radic. Biol. Med. 2019, 134, 119–132. [Google Scholar] [CrossRef]
  51. Wang, X.; Younis, S.; Cen, J.; Wang, Y.; Krizhanovskii, C.; Andersson, L.; Welsh, N. ZBED6 counteracts high-fat diet-induced glucose intolerance by maintaining beta cell area and reducing excess mitochondrial activation. Diabetologia 2021, 64, 2292–2305. [Google Scholar] [CrossRef]
  52. Nishida, Y.; Nawaz, A.; Kado, T.; Takikawa, A.; Igarashi, Y.; Onogi, Y.; Wada, T.; Sasaoka, T.; Yamamoto, S.; Sasahara, M.; et al. Astaxanthin stimulates mitochondrial biogenesis in insulin resistant muscle via activation of AMPK pathway. J. Cachex-Sarcopenia Muscle 2020, 11, 241–258. [Google Scholar] [CrossRef] [Green Version]
  53. Xiao, T.; Liang, X.; Liu, H.; Zhang, F.; Meng, W.; Hu, F. Mitochondrial stress protein HSP60 regulates ER stress-induced hepatic lipogenesis. J. Mol. Endocrinol. 2020, 64, 67–75. [Google Scholar] [CrossRef] [Green Version]
  54. Wen, Z.; Li, J.; Fu, Y.; Zheng, Y.; Ma, M.; Wang, C. Hypertrophic Adipocyte–Derived Exosomal miR-802-5p Contributes to Insulin Resistance in Cardiac Myocytes Through Targeting HSP60. Obesity 2020, 28, 1932–1940. [Google Scholar] [CrossRef]
  55. Kim, K.H.; Jeong, Y.T.; Oh, H.; Kim, S.H.; Cho, J.M.; Kim, Y.-N.; Kim, S.S.; Kim, D.H.; Hur, K.Y.; Kim, H.K.; et al. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat. Med. 2012, 19, 83–92. [Google Scholar] [CrossRef] [Green Version]
  56. Forsström, S.; Jackson, C.B.; Carroll, C.J.; Kuronen, M.; Pirinen, E.; Pradhan, S.; Marmyleva, A.; Auranen, M.; Kleine, I.-M.; Khan, N.A.; et al. Fibroblast Growth Factor 21 Drives Dynamics of Local and Systemic Stress Responses in Mitochondrial Myopathy with mtDNA Deletions. Cell Metab. 2019, 30, 1040–1054.e1047. [Google Scholar] [CrossRef]
  57. Lehtonen, J.M.; Auranen, M.; Darin, N.; Sofou, K.; Bindoff, L.; Hikmat, O.; Uusimaa, J.; Vieira, P.; Tulinius, M.; Lönnqvist, T.; et al. Diagnostic value of serum biomarkers FGF21 and GDF15 compared to muscle sample in mitochondrial disease. J. Inherit. Metab. Dis. 2020, 44, 469–480. [Google Scholar] [CrossRef]
  58. Lee, H.-Y.; Nga, H.T.; Tian, J.; Yi, H.-S. Mitochondrial Metabolic Signatures in Hepatocellular Carcinoma. Cells 2021, 10, 1901. [Google Scholar] [CrossRef]
  59. Ost, M.; Coleman, V.; Voigt, A.; van Schothorst, E.M.; Keipert, S.; van der Stelt, I.; Ringel, S.; Graja, A.; Ambrosi, T.; Kipp, A.P.; et al. Muscle mitochondrial stress adaptation operates independently of endogenous FGF21 action. Mol. Metab. 2015, 5, 79–90. [Google Scholar] [CrossRef]
  60. Park, J.; Joe, Y.; Ryter, S.W.; Surh, Y.-J.; Chung, H.T. Similarities and Distinctions in the Effects of Metformin and Carbon Monoxide in Immunometabolism. Mol. Cells 2019, 42, 292–300. [Google Scholar] [CrossRef]
  61. Moon, J.S.; Goeminne, L.; Kim, J.T.; Tian, J.W.; Kim, S.-H.; Nga, H.T.; Kang, S.G.; Kang, B.E.; Byun, J.; Lee, Y.; et al. Growth differentiation factor 15 protects against the aging-mediated systemic inflammatory response in humans and mice. Aging Cell 2020, 19, e13195. [Google Scholar] [CrossRef] [PubMed]
  62. Choi, M.J.; Jung, S.-B.; Lee, S.E.; Kang, S.G.; Lee, J.H.; Ryu, M.J.; Chung, H.K.; Chang, J.Y.; Kim, Y.K.; Hong, H.J.; et al. An adipocyte-specific defect in oxidative phosphorylation increases systemic energy expenditure and protects against diet-induced obesity in mouse models. Diabetologia 2020, 63, 837–852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yi, H.-S. Implications of Mitochondrial Unfolded Protein Response and Mitokines: A Perspective on Fatty Liver Diseases. Endocrinol. Metab. 2019, 34, 39–46. [Google Scholar] [CrossRef] [PubMed]
  64. Hariyanto, T.I.; Kurniawan, A. Appetite problem in cancer patients: Pathophysiology, diagnosis, and treatment. Cancer Treat. Res. Commun. 2021, 27, 100336. [Google Scholar] [CrossRef]
  65. Rosenberg, B.J.; Hirano, M.; Quinzii, C.M.; Colantuoni, E.; Needham, D.M.; Lederer, D.J.; Baldwin, M.R. Growth differentiation factor-15 as a biomarker of strength and recovery in survivors of acute respiratory failure. Thorax 2019, 74, 1099–1101. [Google Scholar] [CrossRef]
  66. Dominguez-Gonzalez, C.; Badosa, C.; Madruga-Garrido, M.; Martí, I.; Paradas, C.; Ortez, C.; Diaz-Manera, J.; Berardo, A.; Alonso-Pérez, J.; Trifunov, S.; et al. Growth Differentiation Factor 15 is a potential biomarker of therapeutic response for TK2 deficient myopathy. Sci. Rep. 2020, 10, 10111. [Google Scholar] [CrossRef]
  67. Poulsen, N.S.; Madsen, K.L.; Hornsyld, T.M.; Eisum, A.-S.V.; Fornander, F.; Buch, A.E.; Stemmerik, M.G.; Ruiz-Ruiz, C.; Krag, T.; Vissing, J. Growth and differentiation factor 15 as a biomarker for mitochondrial myopathy. Mitochondrion 2020, 50, 35–41. [Google Scholar] [CrossRef]
  68. Montero, R.; Yubero, D.; Villarroya, J.; Henares, D.; Jou, C.; Rodríguez, M.A.; Ramos, F.; Nascimento, A.; Ortez, C.I.; Campistol, J.; et al. GDF-15 Is Elevated in Children with Mitochondrial Diseases and Is Induced by Mitochondrial Dysfunction. PLoS ONE 2016, 11, e0148709. [Google Scholar] [CrossRef] [Green Version]
  69. Straub, I.R.; Weraarpachai, W.; Shoubridge, E.A. Multi-OMICS study of a CHCHD10 variant causing ALS demonstrates metabolic rewiring and activation of endoplasmic reticulum and mitochondrial unfolded protein responses. Hum. Mol. Genet. 2021, 30, 687–705. [Google Scholar] [CrossRef]
  70. Steffen, J.; Ngo, J.; Wang, S.-P.; Williams, K.; Kramer, H.F.; Ho, G.; Rodriguez, C.; Yekkala, K.; Amuzie, C.; Bialecki, R.; et al. The mitochondrial fission protein Drp1 in liver is required to mitigate NASH and prevents the activation of the mitochondrial ISR. Mol. Metab. 2022, 64, 101566. [Google Scholar] [CrossRef]
  71. Aguilar-Recarte, D.; Barroso, E.; Zhang, M.; Rada, P.; Pizarro-Delgado, J.; Peña, L.; Palomer, X.; Valverde, Á.M.; Wahli, W.; Vázquez-Carrera, M. A positive feedback loop between AMPK and GDF15 promotes metformin antidiabetic effects. Pharmacol. Res. 2023, 187, 106578. [Google Scholar] [CrossRef]
  72. Zhang, S.-Y.; Bruce, K.; Danaei, Z.; Li, R.J.; Barros, D.R.; Kuah, R.; Lim, Y.-M.; Mariani, L.H.; Cherney, D.Z.; Chiu, J.F.; et al. Metformin triggers a kidney GDF15-dependent area postrema axis to regulate food intake and body weight. Cell Metab. 2023, 35, 875–886.e5. [Google Scholar] [CrossRef]
  73. Chung, H.K.; Ryu, D.; Kim, K.S.; Chang, J.Y.; Kim, Y.K.; Yi, H.-S.; Kang, S.G.; Choi, M.J.; Lee, S.E.; Jung, S.-B.; et al. Growth differentiation factor 15 is a myomitokine governing systemic energy homeostasis. J. Cell Biol. 2017, 216, 149–165. [Google Scholar] [CrossRef]
  74. Akazawa, S.; Sun, F.; Ito, M.; Kawasaki, E.; Eguchi, K. Efficacy of troglitazone on body fat distribution in type 2 diabetes. Diabetes Care 2000, 23, 1067–1071. [Google Scholar] [CrossRef] [Green Version]
  75. Ensuring drug safety: Lessons from the thiazolidinediones. Lancet 2007, 370, 1101. [CrossRef]
  76. Barber, T.M.; Kabisch, S.; Randeva, H.S.; Pfeiffer, A.F.H.; Weickert, M.O. Implications of Resveratrol in Obesity and Insulin Resistance: A State-of-the-Art Review. Nutrients 2022, 14, 2870. [Google Scholar] [CrossRef]
  77. Zhu, X.; Wu, C.; Qiu, S.; Yuan, X.; Li, L. Effects of resveratrol on glucose control and insulin sensitivity in subjects with type 2 diabetes: Systematic review and meta-analysis. Nutr. Metab. 2017, 14, 60. [Google Scholar] [CrossRef]
  78. Ghorbani, Z.; Hekmatdoost, A.; Mirmiran, P. Anti-Hyperglycemic and Insulin Sensitizer Effects of Turmeric and Its Principle Constituent Curcumin. Int. J. Endocrinol. Metab. 2014, 12, e18081. [Google Scholar] [CrossRef] [Green Version]
  79. Na, L.-X.; Zhang, Y.-L.; Li, Y.; Liu, L.-Y.; Li, R.; Kong, T.; Sun, C.-H. Curcumin improves insulin resistance in skeletal muscle of rats. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 526–533. [Google Scholar] [CrossRef]
  80. Pirola, L.; Balcerczyk, A.; Okabe, J.; El-Osta, A. Epigenetic phenomena linked to diabetic complications. Nat. Rev. Endocrinol. 2010, 6, 665–675. [Google Scholar] [CrossRef]
  81. Zhong, Y.; Xiao, Y.; Gao, J.; Zheng, Z.; Zhang, Z.; Yao, L.; Li, D. Curcumin improves insulin sensitivity in high-fat diet-fed mice through gut microbiota. Nutr. Metab. 2022, 19, 76. [Google Scholar] [CrossRef] [PubMed]
  82. Kim, Y.; Keogh, J.B.; Clifton, P.M. Polyphenols and Glycemic Control. Nutrients 2016, 8, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Thirone, A.C.; Huang, C.; Klip, A. Tissue-specific roles of IRS proteins in insulin signaling and glucose transport. Trends Endocrinol. Metab. 2006, 17, 72–78. [Google Scholar] [CrossRef] [PubMed]
  84. Jayanthy, G.; Devi, V.R.; Ilango, K.; Subramanian, S.P. Rosmarinic Acid Mediates Mitochondrial Biogenesis in Insulin Resistant Skeletal Muscle Through Activation of AMPK. J. Cell. Biochem. 2017, 118, 1839–1848. [Google Scholar] [CrossRef] [PubMed]
  85. Hartogh, D.J.D.; Vlavcheski, F.; Tsiani, E. Muscle Cell Insulin Resistance Is Attenuated by Rosmarinic Acid: Elucidating the Mechanisms Involved. Int. J. Mol. Sci. 2023, 24, 5094. [Google Scholar] [CrossRef]
  86. Mimura, J.; Inose-Maruyama, A.; Taniuchi, S.; Kosaka, K.; Yoshida, H.; Yamazaki, H.; Kasai, S.; Harada, N.; Kaufman, R.J.; Oyadomari, S.; et al. Concomitant Nrf2- and ATF4-Activation by Carnosic Acid Cooperatively Induces Expression of Cytoprotective Genes. Int. J. Mol. Sci. 2019, 20, 1706. [Google Scholar] [CrossRef] [Green Version]
  87. Santana, L.F.; Inada, A.C.; Santo, B.L.S.D.E.; Filiú, W.F.O.; Pott, A.; Alves, F.M.; Guimarães, R.d.C.A.; Freitas, K.d.C.; Hiane, P.A. Nutraceutical Potential of Carica papaya in Metabolic Syndrome. Nutrients 2019, 11, 1608. [Google Scholar] [CrossRef] [Green Version]
  88. Elsayed, R.H.; Kamel, E.M.; Mahmoud, A.M.; El-Bassuony, A.A.; Bin-Jumah, M.; Lamsabhi, A.M.; Ahmed, S.A. Rumex dentatus L. phenolics ameliorate hyperglycemia by modulating hepatic key enzymes of carbohydrate metabolism, oxidative stress and PPARγ in diabetic rats. Food Chem. Toxicol. 2020, 138, 111202. [Google Scholar] [CrossRef]
  89. Song, P.; Kim, J.H.; Ghim, J.; Yoon, J.H.; Lee, A.; Kwon, Y.; Hyun, H.; Moon, H.-Y.; Choi, H.-S.; Berggren, P.-O.; et al. Emodin Regulates Glucose Utilization by Activating AMP-activated Protein Kinase. J. Biol. Chem. 2013, 288, 5732–5742. [Google Scholar] [CrossRef] [Green Version]
  90. Yu, F.; Yu, N.; Peng, J.; Zhao, Y.; Zhang, L.; Wang, X.; Xu, X.; Zhou, J.; Wang, F. Emodin inhibits lipid accumulation and inflammation in adipose tissue of high-fat diet-fed mice by inducing M2 polarization of adipose tissue macrophages. FASEB J. 2021, 35, e21730. [Google Scholar] [CrossRef]
  91. Fu, S.; Dang, Y.; Xu, H.; Li, A.; Zhou, X.; Gao, X.; Li, Z. Aloe vera-Fermented Beverage Ameliorates Obesity and Gut Dysbiosis in High-Fat-Diet Mice. Foods 2022, 11, 3728. [Google Scholar] [CrossRef]
  92. Dou, F.; Liu, M.Y.; Liu, L.; Wang, J.; Sun, M.T.; Mu, M.F.; Guo, Q.; Guo, C.; Jia, N.; Liu, W.; et al. Aloe-Emodin Ameliorates Renal Fibrosis Via Inhibiting PI3K/Akt/mTOR Signaling Pathway In Vivo and In Vitro. Rejuvenation Res. 2019, 22, 218–229. [Google Scholar] [CrossRef] [Green Version]
  93. Quan, Y.; Gong, L.; He, J.; Zhou, Y.; Liu, M.; Cao, Z.; Li, Y.; Peng, C. Aloe emodin induces hepatotoxicity by activating NF-κB inflammatory pathway and P53 apoptosis pathway in zebrafish. Toxicol. Lett. 2019, 306, 66–79. [Google Scholar] [CrossRef]
  94. Régnier, M.; Rastelli, M.; Morissette, A.; Suriano, F.; Le Roy, T.; Pilon, G.; Delzenne, N.M.; Marette, A.; Van Hul, M.; Cani, P.D. Rhubarb Supplementation Prevents Diet-Induced Obesity and Diabetes in Association with Increased Akkermansia muciniphila in Mice. Nutrients 2020, 12, 2932. [Google Scholar] [CrossRef]
  95. Zhou, Y.; Gao, C.; Vong, C.T.; Tao, H.; Li, H.; Wang, S.; Wang, Y. Rhein regulates redox-mediated activation of NLRP3 inflammasomes in intestinal inflammation through macrophage-activated crosstalk. Br. J. Pharmacol. 2022, 179, 1978–1997. [Google Scholar] [CrossRef]
  96. Genua, M.; Rutella, S.; Correale, C.; Danese, S. The triggering receptor expressed on myeloid cells (TREM) in inflammatory bowel disease pathogenesis. J. Transl. Med. 2014, 12, 293. [Google Scholar] [CrossRef] [Green Version]
  97. Wang, Q.-Y.; Tong, A.-H.; Pan, Y.-Y.; Zhang, X.-D.; Ding, W.-Y.; Xiong, W. The effect of cassia seed extract on the regulation of the LKB1–AMPK–GLUT4 signaling pathway in the skeletal muscle of diabetic rats to improve the insulin sensitivity of the skeletal muscle. Diabetol. Metab. Syndr. 2019, 11, 1–10. [Google Scholar] [CrossRef] [Green Version]
  98. Zhou, F.; Ding, M.; Gu, Y.; Fan, G.; Liu, C.; Li, Y.; Sun, R.; Wu, J.; Li, J.; Xue, X.; et al. Aurantio-Obtusin Attenuates Non-Alcoholic Fatty Liver Disease Through AMPK-Mediated Autophagy and Fatty Acid Oxidation Pathways. Front. Pharmacol. 2021, 12, 826628. [Google Scholar] [CrossRef]
  99. Guo, C.; Liao, W.; Qiu, R.; Zhou, D.; Ni, W.; Yu, C.; Zeng, Y. Aurantio-obtusin improves obesity and insulin resistance induced by high-fat diet in obese mice. Phytother. Res. 2020, 35, 346–360. [Google Scholar] [CrossRef] [PubMed]
  100. Jung, H.A.; Ali, M.Y.; Choi, J.S. Promising Inhibitory Effects of Anthraquinones, Naphthopyrone, and Naphthalene Glycosides, from Cassia obtusifolia on α-Glucosidase and Human Protein Tyrosine Phosphatases 1B. Molecules 2017, 22, 28. [Google Scholar] [CrossRef] [Green Version]
  101. Patel, D.; Prasad, S.; Kumar, R.; Hemalatha, S. An overview on antidiabetic medicinal plants having insulin mimetic property. Asian Pac. J. Trop. Biomed. 2012, 2, 320–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Zhai, B.; Zhang, C.; Sheng, Y.; Zhao, C.; He, X.; Xu, W.; Huang, K.; Luo, Y. Hypoglycemic and hypolipidemic effect of S-allyl-cysteine sulfoxide (alliin) in DIO mice. Sci. Rep. 2018, 8, 3527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. El-Saber Batiha, G.; Magdy Beshbishy, A.; Wasef, L.G.; Elewa, Y.H.; Al-Sagan, A.A.; El-Hack, A.; Taha, M.E.; Abd-Elhakim, Y.M.; Prasad Devkota, H. Chemical Constituents and Pharmacological Activities of Garlic (Allium sativum L.): A Review. Nutrients 2020, 12, 872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Faroughi, F.; Mohammad-Alizadeh-Charandabi, S.; Javadzadeh, Y.; Mirghafourvand, M. Effects of Garlic Pill on Blood Glucose Level in Borderline Gestational Diabetes Mellitus: A Randomized Controlled Trial. Iran. Red Crescent Med. J. 2018, 20, e60675. [Google Scholar] [CrossRef]
  105. Pivari, F.; Mingione, A.; Brasacchio, C.; Soldati, L. Curcumin and Type 2 Diabetes Mellitus: Prevention and Treatment. Nutrients 2019, 11, 1837. [Google Scholar] [CrossRef] [Green Version]
  106. Zhou, T.; Gai, Z.; Gao, X.; Li, L. The Potential Mechanism of Exercise Combined with Natural Extracts to Prevent and Treat Postmenopausal Osteoporosis. J. Health Eng. 2021, 2021, 2852661. [Google Scholar] [CrossRef]
  107. Wieërs, G.; Verbelen, V.; Driessche, M.V.D.; Melnik, E.; Vanheule, G.; Marot, J.-C.; Cani, P.D. Do Probiotics During In-Hospital Antibiotic Treatment Prevent Colonization of Gut Microbiota with Multi-Drug-Resistant Bacteria? A Randomized Placebo-Controlled Trial Comparing Saccharomyces to a Mixture of Lactobacillus, Bifidobacterium, and Saccharomyces. Front. Public Health 2021, 8, 578089. [Google Scholar] [CrossRef]
  108. Leyva-Gómez, G.; González-Torres, M.; Alcalá-Alcalá, S.; Bernal-Chávez, S.A.; Morales-Morfin, J.C.; Carmen, M.G.-D.; Sharifi-Rad, J.; Figueroa-González, G.; Reyes-Hernández, O.D.; Del Prado-Audelo, M.L.; et al. Development of films from natural sources for infections during wound healing. Cell. Mol. Biol. 2021, 67, 96–100. [Google Scholar] [CrossRef]
  109. Bellik, Y.; Hammoudi, S.M.; Abdellah, F.; Iguer-Ouada, M.; Boukraâ, L. Phytochemicals to prevent inflammation and allergy. Recent Patents Inflamm. Allergy Drug Discov. 2012, 6, 147–158. [Google Scholar] [CrossRef]
Figure 1. Extensive use and specific mechanisms of natural drugs in insulin resistance-related diseases. The arrows between the sections represent the preventive and therapeutic effects of the natural drug on the disease through the factors shown next to the arrows.
Figure 1. Extensive use and specific mechanisms of natural drugs in insulin resistance-related diseases. The arrows between the sections represent the preventive and therapeutic effects of the natural drug on the disease through the factors shown next to the arrows.
Molecules 28 05525 g001
Figure 2. The relevant factors mentioned in this review that affect mitochondrial stress and insulin resistance or connect the two. Some pathological factors lead to increased mitochondrial stress in the body. This process can be induced or aggravated by factors such as cyclophilin D (CypD) and advanced glycation end products (AGEs), but newly discovered factors such as peroxisome proliferator-activated receptors (PPARs) and proliferator-activated receptor-γ coactivator-1 (PGC-1) can improve this pathological process. Among miRNAs, miR-30c can inhibit mitochondrial stress and insulin resistance by targeting PGC-1, miR-126 can protect the body from the effects of reactive oxygen species (ROS) imbalance, and miR-155-5p can improve the adaptation of cells to insulin resistance. Mitochondrial stress leads to an increase in ROS production, which aggravates insulin resistance in many ways, such as the overexpression of heat shock protein 60 (HSP60). However, mitochondrial stress does not only cause purely negative effects. Its activation of fibroblast growth factor 21 (FGF21) can inhibit the progression of insulin resistance to a certain extent, and we hypothesize that its activation of growth differentiation factor-15 (GDF-15) may do the same. Solid arrows indicate “direct action” with no other processes in between; The dashed arrow represents “indirect action”, which means that there are multiple action processes that have been omitted.
Figure 2. The relevant factors mentioned in this review that affect mitochondrial stress and insulin resistance or connect the two. Some pathological factors lead to increased mitochondrial stress in the body. This process can be induced or aggravated by factors such as cyclophilin D (CypD) and advanced glycation end products (AGEs), but newly discovered factors such as peroxisome proliferator-activated receptors (PPARs) and proliferator-activated receptor-γ coactivator-1 (PGC-1) can improve this pathological process. Among miRNAs, miR-30c can inhibit mitochondrial stress and insulin resistance by targeting PGC-1, miR-126 can protect the body from the effects of reactive oxygen species (ROS) imbalance, and miR-155-5p can improve the adaptation of cells to insulin resistance. Mitochondrial stress leads to an increase in ROS production, which aggravates insulin resistance in many ways, such as the overexpression of heat shock protein 60 (HSP60). However, mitochondrial stress does not only cause purely negative effects. Its activation of fibroblast growth factor 21 (FGF21) can inhibit the progression of insulin resistance to a certain extent, and we hypothesize that its activation of growth differentiation factor-15 (GDF-15) may do the same. Solid arrows indicate “direct action” with no other processes in between; The dashed arrow represents “indirect action”, which means that there are multiple action processes that have been omitted.
Molecules 28 05525 g002
Figure 3. The current main applications of GDF-15 and its role in mitochondrial stress and insulin resistance. GDF-15 is an important marker in the diagnosis of many diseases. In addition, it can be used to treat the abnormal pathological state of mitochondrial stress, which is elevated to varying degrees in a variety of diseases, and its expression can effectively improve insulin resistance. For example, by activating ATF4, metformin effectively alleviates insulin resistance by inducing GDF-15. Notably, CypD may aggravate the progression of diseases related to insulin resistance by inhibiting the expression of GDF-15.
Figure 3. The current main applications of GDF-15 and its role in mitochondrial stress and insulin resistance. GDF-15 is an important marker in the diagnosis of many diseases. In addition, it can be used to treat the abnormal pathological state of mitochondrial stress, which is elevated to varying degrees in a variety of diseases, and its expression can effectively improve insulin resistance. For example, by activating ATF4, metformin effectively alleviates insulin resistance by inducing GDF-15. Notably, CypD may aggravate the progression of diseases related to insulin resistance by inhibiting the expression of GDF-15.
Molecules 28 05525 g003
Table 1. A brief summary of the relevant mechanisms mentioned in the review.
Table 1. A brief summary of the relevant mechanisms mentioned in the review.
FactorMechanism
AGEsPromotes mitochondrial stress by activating NADPH
CypDPromotes mitochondrial stress when the MPTP is open and NEFAs are present
PPARsInhibit mitochondrial stress by inhibiting IL-6
miR-126Inhibits mitochondrial stress by activating SIRT1 and SOD
miR-30cInhibits mitochondrial stress by activating PGC-1 and PPAR-α
HSP60Mitochondrial stress causes overexpression of HSP60; HSP60 promotes mTORC1-SREBP1 signal transduction and promotes insulin resistance together with endoplasmic reticulum stress
FGF21Metformin induces mitochondrial stress through the Perk-eIF2α-ATF4 axis; mitochondrial stress promotes FGF21 expression by activating the comprehensive stress response (ISR); FGF21 improves insulin resistance
GDF-15GDF-15 improves insulin resistance; CYPD may cause insulin resistance by inhibiting GDF-15
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wu, P.; Wang, X. Natural Drugs: A New Direction for the Prevention and Treatment of Diabetes. Molecules 2023, 28, 5525. https://doi.org/10.3390/molecules28145525

AMA Style

Wu P, Wang X. Natural Drugs: A New Direction for the Prevention and Treatment of Diabetes. Molecules. 2023; 28(14):5525. https://doi.org/10.3390/molecules28145525

Chicago/Turabian Style

Wu, Peishan, and Xiaolei Wang. 2023. "Natural Drugs: A New Direction for the Prevention and Treatment of Diabetes" Molecules 28, no. 14: 5525. https://doi.org/10.3390/molecules28145525

Article Metrics

Back to TopTop