Yongting Zhao, Xiaofang Zhang, Haihai Liang, Lihong Wang. Antidiabetic agents: Do they hit the right targets?[J]. Frigid Zone Medicine, 2022, 2(4): 225-243. doi: 10.2478/fzm-2022-0030
Citation: Yongting Zhao, Xiaofang Zhang, Haihai Liang, Lihong Wang. Antidiabetic agents: Do they hit the right targets?[J]. Frigid Zone Medicine, 2022, 2(4): 225-243. doi: 10.2478/fzm-2022-0030

Antidiabetic agents: Do they hit the right targets?

doi: 10.2478/fzm-2022-0030
More Information
  • Corresponding author: Lihong Wang, E-mail: nd6688@163.com
  • Received Date: 2021-07-09
  • Accepted Date: 2022-04-21
  • Available Online: 2022-10-30
  • Diabetes mellitus (DM) is a progressive metabolic disease characterized by chronic hyperglycemia and caused by different degree of pancreatic islet dysfunction and/or insulin resistance (IR). Long course DM can lead to a variety of macrovascular and microvascular complications which involve artery vessels, heart, kidney, retina, nervous system, etc. In recent years, DM has attracted more and more attention due to its high morbidity and mortality. In addition to achieve effective glycemic control, prevention of complications has also been considered a priority for type 2 diabetes mellitus (T2DM) management. Herein, we provide a comprehensive overview on the pharmacotherapeutics for T2DM and perspectives on the future directions of basic and translational research on anti-diabetic therapy and pharmatheutical development of new drugs.

     

  • Diabetes mellitus (DM) is a progressive non-communicable metabolic disease characterized by chronic hyperglycemia and pancreatic islet dysfunction, which eventually develops into a variety of severe macrovascular and microvascular complications, causing tremendous premature morbidity and mortality[1-3]. According to statistics by the International Diabetes Federation, diabetes caused 6.7 million deaths in 2021, and 537 million adults aged between 20 and 79 years old were living with diabetes; yet this number will likely rise up to 783 million by 2045. DM, particularly type 2 diabetes mellitus (T2DM), constitutes one of the most critical public health challenges of the twenty-first century, with enormous and continuous increasing economic and social burdens due to its high prevalence, severe co-morbidities, negative impact on quality of life, and life-threatening complications. Even worse is the prevalence of T2DM has reached approximately 10% of the global population and keeps rising rapidly worldwide. Asia has become the major region of emerging T2DM incidence with China and India being the top two epicenters[1-4]. As a metabolic disorder, the current global epidemic of T2DM is mainly driven by an unhealthy diet and a sedentary lifestyle, although genetic predisposition partly determines individual susceptibility to T2DM. At present, T2DM is primarily treated by the pharmacologic approach aiming to control blood glucose and minimize/postpone the associated complications. Anti-diabetic drugs are increasingly used worldwide, and the therapeutics has been continuously evolving with emerging new agents. Yet, the currently available anti-diabetic drugs do not present the desired efficacy but are generally associated with serious adverse effects. Herein, we intend to provide a comprehensive overview on the pharmacological therapy of T2DM and perspectives on the future directions of pharmacological research on anti-diabetic agents. Specifically, we summarize the published data with most updated information about our current understanding of the cell/molecular mechanisms for T2DM, the efficacy and mechanisms of various categories of anti-diabetic agents, the potential applications of some promising natural health products to the treatment of T2DM, and some of the novel anti-diabetic agents under development (undergoing clinical trials). We provide a balanced review and critical thoughts on the findings and discuss the current challenges and opportunities in pharmacological therapy of T2DM.

    Over 90% of DM cases belong to T2DM, and epidemiology of T2DM is affected by multiple non-modifiable and modifiable risk factors of complex combination of genetic, metabolic, and environmental factors that interact with one another[2-3, 5]. The T2DM epidemic can be ascribed primarily to the global rise in obesity, sedentary lifestyles, high caloric diets, and population aging[1]. T2DM is a multi-mechanism and multisystem disease, and therefore management of this pathological entity requires not only precise targeting of the diabetic mechanisms but also satisfactory avoidance of various complications especially cardiovascular complication. The prerequisite to achieve these goals is to have better understanding the mechanisms of T2DM (Fig. 1).

    Figure  1.  T2DM is a multi-mechanism and multisystem disease, involving β-cell dysfunction, IR, dyslipidemia, inflammation, gut microbiota, mitochondrial dysfunction, cardiovascular complication and metabolic memory and diabetic complications
    IR, insulin resistance; T2DM, type 2 diabetes mellitus.

    T2DM is a metabolic condition marked by deficient insulin secretion by pancreatic islet β-cells and tissue insulin resistance (IR)[2-3], which is manifested by loss of glucose homeostasis or hyperglycemia. In case of β-cell dysfunction, insulin secretion is reduced, which profoundly limits the body's capacity to maintain physiological glucose levels. β-cell dysfunction has been traditionally associated with β-cell death[6]. However, recent studies support that β-cell dysfunction in T2DM is consequent to a more complex network of interactions between the environment and various molecular pathways[7]. In obese population or subjects in an excessive nutritional state, hyperglycemia and hyperlipidemia are common characteristics that often cause IR and chronic inflammation. Such circumstances render β-cells under great toxic pressures created by lipotoxicity, glucotoxicity and glucolipotoxicity, which in turn result in inflammation, inflammatory stress, endoplasmic reticulum (ER) stress, metabolic/oxidative stress, and amyloid stress, ultimately leading to a loss of islet integrity thereby β-cell dysfunction with deficient insulin secretion [6-7].

    In regard with IR, however, insulin secretion may or may not be impacted, but glucose production in liver is increased whereas glucose uptake in muscle, liver, and adipose tissue is decreased. IR refers to a decrease in metabolic response of insulin-responsive cells to insulin or, at a systemic level, an impaired/lower response of cells/tissues/organs to circulating insulin[8-9]. In fasting state, insulin response is diminished by glucagon, glucocorticoids and catecholamines in order to prevent insulin-induced hypoglycemia. Catecholamines enhance lipolysis and glycogenolysis, whereas glucocorticoids promote muscle catabolism, gluconeogenesis and lipolysis. Hence, excessive secretion of these hormones is believed to account for the induction of IR[5, 8-10]. Three main extra-pancreatic insulin-sensitive organs, skeletal muscle, adipose tissue, and liver, play major roles in the development of systemic IR, leading to T2DM.

    In many cases, both β-cell dysfunction and IR processes take place together in the pathogenesis and development of T2DM, and the decrease of β-cell function gradually aggravates with the course of disease. Under such a circumstance, hyperglycemia is amplified leading to the progression of T2DM[11-12].

    Dyslipidemia is a common feature of T2DM, and increases the incidence of atherosclerosis and mortality of diabetic patients[13]. The hallmark of diabetic dyslipidemia is a characteristic dyslipidemia profile consisting of elevated triglyceride (TG), TG-rich lipoproteins, small dense low-density lipoproteins (LDLs), and reduced high-density lipoprotein (HDL) levels[14]. Although the pathophysiology of dyslipidemia in T2DM is still incompletely characterized, several factors such as hyperglycemia, IR, hyperinsulinemia, abnormalities in adipokines and adipocytokines have been implicated[15]. Epidemiological studies indicate that TG-rich lipoproteins contribute to atherogenesis and cardiovascular disease (CVD) risk[16-17].

    Sustained high glucose levels increase proinsulin biosynthesis and islet amyloid polypeptides (IAAP) in β-cells, leading to the accumulation of misfolded insulin and IAAP and increase in the production of oxidative protein folding-mediated reactive oxygen species (ROS)[18]. These effects alter physiological ER Ca2+ mobilization and favor proapoptotic signals, proinsulin mRNA degradation and induce interleukin (IL)-1 β release that recruits macrophages and enhances local islet inflammation[7].

    Gut microbiota is composed of many microbial species and recent evidence indicates that gut microbiota tremendously impact human physiology and participate in different biological processes[19]. A high-fat diet can induce up to threefold production of lipopolysaccharide (from Gram-negative bacteria) in mice models, thereby contributing to low-grade inflammation and IR[20]. Furthermore, intestinal dysbiosis can reduce short-chain fatty acid synthesis that promotes gut barrier integrity, pancreatic β-cell proliferation and insulin biosynthesis[21]. Dysbiosis can also compromise the production of other metabolites such as branched amino-acids and trimethylamine thus disrupting glucose homeostasis and triggering T2DM development.

    Evidence is emerging linking mitochondrial dysfunction to T2DM, age-related IR and associated complications[22-23]. Many damaging factors such as oxidative stress, defective mitochondrial biogenesis, genetic mutations, and aging can disrupt mitochondrial integrity causing mitochondrial dysfunction. Deregulated mitochondrial homeostasis with reduced mitochondrial biogenesis is a key component of mitochondrial dysfunction. The damage induced by excessive oxidative stress in mitochondria activates mitophagic processes in order to eliminate dysfunctional mitochondria or apoptotic cells[24]. The role of mitochondrial genetics is another risk of T2DM with several mitochondrial DNA (mtDNA) variants being associated with T2DM development[25-27].

    T2DM is a multisystem disease with a strong correlation with CVD that is the major diabetic complication[28]. T2DM leads to a two- to four-fold increase in the mortality rate of adults from heart disease and stroke and is associated with both micro-and macro-vascular complications, with the latter accelerating atherosclerosis leading to severe peripheral vascular disease, premature coronary artery disease and increased risk of cerebrovascular diseases[28-30].

    Glycemic control is an important strategy in management of DM. However, the results of multiple large-scale clinical trials revealed that even with efficient glycemic control, diabetic complications persist, which is referred as "metabolic memory". This concept arose from the results of multiple large-scale clinical trials, which showed that even years before the diagnosis of diabetes, diabetes complications have already existed and progressed even when glycemic control is restored through pharmacological intervention[31-32]. Animal models of diabetes and in vitro cell cultures also subsequently confirmed that the initial hyperglycemic period results in permanent abnormalities (including aberrant gene expression) of target organs/cells[33-34]. It is now believed that metabolic memory involves four mechanisms: epigenetics[35-38], oxidative stress[39-40], non-enzymatic glycation of proteins and chronic inflammation.

    In addition, low-grade inflammation, which is known to contribute importantly to T2DM and its vascular complications, has been shown to mediate metabolic memory. Many environmental factors (age, obesity, sedentarism and diet) that promote T2DM development trigger an inflammatory response leading to IR and endothelial dysfunction[41-42]. NF-κB activation in T2DM can induce the expression of inflammatory genes, which in turn enhances monocyte binding to endothelial and vascular smooth muscle cells, and subsequently promoting monocyte-to-macrophage differentiation[41]. In addition, NF-κB can also induces expression of inflammatory cytokines that are involved in vascular inflammation, with subsequent generation of endothelial adhesion molecules, proteases, and other mediators[42].

    In theory, each of the T2DM-underlying mechanisms could be a therapeutic target for anti-diabetic drugs. In reality, however, as we can see from the following section, currently available anti-diabetic agents in clinical use mainly focus on a few of the multimechanisms for T2DM.

    At present, hypoglycemic drugs that are commonly used in clinic are divided into nine categories, including biguanides, sulfonylureas, glitinides, thiazolidinediones, α-glucosidase inhibitors, dipeptidyl-peptidase Ⅳ (DPP-Ⅳ) inhibitors, sodium glucose cotransporter 2 (SGLT2) inhibitors, glucagon-like peptide 1 receptor agonists (GLP-1RAs) and insulin (Table 1). Most of the drugs are administered orally, except for a few of them, such as insulin and GLP-1RAs. There are other anti-diabetic drugs that are not commonly used in clinic, such as PPAR agonist, protein tyrosine phosphatase 1B (PTP1B) inhibitors, aldose reductase inhibitors (ARIs), α-glucosidase inhibitors and G protein-coupled receptor (GPCR) agonists (Table 2).

    Table  1.  Contemporary anti-diabetic drugs
    Hypoglycemic drugs Advantages Disadvantage Function Mechanism
    Biguanides The first-line pharmacological choice for T2DM None Suppressing hepatic gluconeogenesis and glucose output and promoting the uptake and utilization of glucose Activating AMPK and inhibiting the transcription of gluconeogenic genes
    Sulfonylureas Specifically treating non-insulin-dependent DM Hypoglycemia and weight gain Stimulating insulin secretion and potentiating the biologic effect of the insulin Binding to the SUR1
    Glitinides (non-sulfonylureas) Display excellent safety and efficacy and have low risk of hypoglycemia The response to glinides varies Among individuals Promoting the release of postprandial insulin Inhibiting KATP in the pancreatic β-cell membrane and inhibiting KCNQ1
    Thiazolidinediones The only pharmacologic agents that specifically treat IR Fluid retention, heart failure, weight gain, peripheral fractures, and macular edema Increasing insulin sensitivity and inhibiting inflammation Activating PPARγ
    α-glucosidase inhibitors Efficacy, safety, tolerability cardiovascular benefits, and lack of hypoglycemia Not suitable candidates for other antidiabetic drugs and abdominal distension Delaying intestinal carbohydrate absorption, Reducing postprandial glycaemia and saving insulin Inhibiting upper gastrointestinal enzymes α-amylase and other α-glucosidases
    Dipeptidyl-peptidase Ⅳ inhibitors Have good security and are worthy of wide application if the indications are appropriate Upper respiratory tract infection, nasopharyngitis, headache, urinary tract infection, arthralgia Increasing insulin secretion and decreasing glucagon secretion Enhancing GLP-1 preservation and expansion of β-cell mass through the inhibition of apoptotic pathways
    Sodium glucose cotransporter 2 inhibitors The combination of metformin and SGL T2 inhibitors Urinary tract and genital infections, euglycemic diabetic ketoacidosis Inhibiting the absorption of glucose from the proximal tubule of the kidney Inhibiting the accumulation of M1-polarized macrophages and affecting inflammatory
    Glucagon-like peptide 1 receptor agonists The only islet G protein-coupled receptors in current clinical Mild side effects of nausea, vomiting and hypersensitivity Promoting insulin secretion of islet β-cells, inhibit appetite and delay gastric emptying Activating GLP-1receptors
    T2DM, type 2 diabetes mellitus; AMPK, adenosine monophosphate-activated protein kinase; KATP, ATP-sensitive potassium channel; PPARγ, peroxisome proliferator-activated receptor γ; GLP-1, glucagon-like peptide 1;PTPs, protein tyrosine phosphatases; IR, insulin resistance; SUR1, sulfonylurea receptors.
     | Show Table
    DownLoad: CSV
    Table  2.  New anti-diabetic drugs
    Hypoglycemic drugs Advantages Disadvantage Function
    Insulin analogues The most effective method of lowering blood glucose Imitating natural insulin with exogenous one remains a challenge Faster insulin absorption
    PTP1B inhibitors Favorable curing effects The structural homologies of PTPs remains a challenge Regulating insulin and leptin signal transduction negatively
    ARIs Applied to diabetic complications Ineffective treatment of diabetes Diminishing sorbitol flux through suppressing the polyol
    G protein-coupled receptor agonists Low risk of inducing hypoglycemia and gaining body weight Failed to yield metabolic benefits in humans with T2DM Enhancing insulin secretion and reduction appetite
    Anti-diabetic phytochemicals Prevention and management of T2DM with dietary polyphenols Nadequate evidence Inhibition of intestinal α-glucosidase and α-amylase, lens aldose reductase, etc
    Resveratrol Naturally occurring phytoalexin Nadequate evidence Reduction of blood glucose, improvement of insulin sensitivity, preservation of pancreatic β-cells, alleviation of diabetic complications
    Curcumin A bioactive constituent Unknow Improving the function and prevention death of β-cells, and decreasing IR
    Berberine An adjuvant treatment of T2DM, hyperlipidemia, and hypertension Unknow Increasing insulin receptor expression
    ARIs, aldose reductase inhibitors; T2DM, type 2 diabetes mellitus; PTP1B, protein tyrosine phosphatase 1B; IR, insulin resistance; PTPs, protein tyrosine phosphatases.
     | Show Table
    DownLoad: CSV

    Biguanides are a class of hypoglycemic drugs of herbal origin, represented by metformin that has been widely used to treat diabetes since the 1950s[43]. Metformin is the first-line pharmacological choice for the management of hyperglycemia in T2DM and improves glycemic control without inducing hypoglycemia or weight gain[44]. The efficacy of metformin has been confirmed by many experiments, one of which is a randomized clinical trial of intensive vs. standard glycemic control among patients with newly diagnosed T2DM[45-46]. Metformin is believed to act by suppressing hepatic gluconeogenesis and glucose output on one hand and promoting the uptake and utilization of glucose in skeletal muscles and adipose tissues to normalize blood glucose level on the other hand, based on numerous animal and human studies[47]. Also important is that metformin has certain insulin-sensitizing effects likely by acting in the gut lumen through multiple mechanisms. Among high risk patients with prediabetes and IR, metformin reduces the risk of developing diabetes[48].

    In addition to its hypoglycemic and insulin-sensitizing effects, metformin also has protective effects on cardiovascular system[45] and beneficial actions on Alzheimer's disease through improving cognitive impairment and memory loss[49]. A clinical trial found lower rates of myocardial infarction and mortality in a subgroup of overweight patients randomly assigned to metformin compared with conventional (diet) therapy[45]. Moreover, metformin can also exert many other favorable effects beyond its glucose-lowering action, among which are anti-inflammatory, anti-cancer, immunomodulatory effects, as well as reduction of free fatty acids and improvement of endothelial dysfunction[50-53]. Evidence also exists indicating that metformin prolongs lifespan by activating autophagy and adenosine monophosphate-activated protein kinase (AMPK) which inhibits mTOR[54]. It should be noted that interest in metformin for other uses remains yet to be supported by solid clinical evidence[47].

    While it appears that metformin has a hundred merits and not a single demerit due to its pleiotropic property, a full understanding of the mechanisms of action remains elusive. Nonetheless, this drug is known to produce its effects primarily by activating AMPK, increasing the absorption of intestinal glucose and the level of GLP-1, and improving gut microbiome[55]. At the molecular level, metformin improves insulin sensitivity by inhibiting the transcription of gluconeogenic genes thereby the expression of the corresponding protein products and lipogenesis[56].

    Because of its multiple effects, metformin has been widely used either alone or in combination with other agents in T2DM patients. Overall, six decades of clinical uses indicate no major safety concerns for metformin, except for subclinical increases in lactic acid or lactic acidosis in extreme overdose or improper selection of indications [46].

    It is known that patients with T2DM is associated with both poorer clinical outcomes during the corona virus disease 2019 (COVID-19) pandemic and an increased risk of death in hospitalized COVID-19 patients. Thus, it is natural and rational to speculate the potential benefit of anti-diabetic agents in improving the prognosis of COVID-19 patients. Indeed, the role of glucose control has been emphasized for such a purpose[57-59]. it may be speculated that this biguanide might positively influence the prognosis of patients with T2DM hospitalized for COVID-19. The available data from observational retrospective studies present strong evidence for significant reduction in mortality in metformin users compared with non-users. A meta-analysis on four retrospective analyses of observational studies in patients with T2DM hospitalized for severe COVID-19 suggests a positive effect (whether statistically significant or not) in all four studies with an overall reduction of death by 25%[60-61]. For example, a large US study of 6 256 COVID-19 patients with T2DM (52.8% female, mean age 75 years) associated metformin with significant reduction of mortality in women, but not in men[62]. These studies indicate the potential novel application of metformin to COVID-19 patients with T2DM for rescuing the lives and meanwhile achieving effective control of glucose on one hand and support the importance of glycemic control to related conditions other than diabetes on the other hand. Nonetheless, given the inherent confounding nature of observational studies, caution must be taken before drawing any firm conclusions in the absence of randomized controlled trials.

    Sulfonylureas are commonly used as insulin secretagogues. Sulfonylurea drugs are usually categorized as being first-, second-generation and vary in terms of their dosages and effectiveness in treating T2DM[63]. The first-generation sulfonylureas mainly include tolbutamide, chlorpropamide, acetohexamide and tolazamide, and the second-generation sulfonylureas such as glyburide, glipizide and glibornuride have a more nonpolar or lipophilic side chain, which results in a marked increase in their hypoglycemic potency. Although controversies exist as to the usefulness of sulfonylureas, evidence is generally in favor of their use in T2DM with poor islet function[64]. When used properly, the sulfonylurea drugs lower plasma glucose concentrations in diabetic patients by stimulating insulin secretion and by potentiating the biologic effect of the insulin on tissues as skeletal muscle, fat, and liver. Moreover, sulfonylureas can reduce the clearance rate of insulin in liver, resulting in increased insulin levels[65], and prevent α cells from secreting glucagon[66]. Furthermore, sulfonylureas can also improve the sensitivity of peripheral tissues to insulin by binding and activating peroxisome proliferator-activated receptor γ (PPARγ) receptor[67] and promote the utilization of peripheral glucose[68]. In addition to their hypoglycemic effect, these drugs can also inhibit the proliferation of vascular smooth muscle cells and prevent the progression of DM-related atherosclerosis[69-71]. The main adverse effect of sulfonylureas is long-lasting hypoglycemia, which may lead to permanent neurological damage and even death in elderly subjects who are exposed to some intercurrent events, e.g., acute energy deprivation or a drug interaction. The first generation of sulfonylureas has severe cardiovascular safety issues, and the second and third generation sulfonylureas do not increase the risk of all-cause mortality, cardiovascular mortality, myocardial infarction, and stroke[72]. However, at present, there is no consensus on cardiovascular safety of sulfonylureas. There is also a hypothesis that sulfonylureas may be related to hypertension by hindering the relaxation of vascular smooth muscle, but it needs to be further confirmed[73]. Although sulfonylureas have side effects such as hypoglycemia and weight gain, they could be used carefully according to patient's conditions.

    Regarding the mechanism of action, sulfonylureas act mainly by binding to the sulfonylurea receptors (SUR1) of ATP-sensitive potassium channel (KATP) in β-cells to inhibit potassium efflux and depolarize cell membrane so as to stimulate insulin secretion in a glucose-independent manner[74]. Some of the first generation sulfonylureas can also interact with the sulfonylurea receptors in cardiomyocytes (SUR2A) and vascular smooth muscle (SUR2B) cells, resulting in the disturbance of cardiac conduction in myocardial infarction[75]. Through a similar mechanism, sulfonylureas stimulate the secretion of somatostatin and suppress the secretion of glucagon in δ-cells and α-cells[76-77]. Sulfonylureas seem to exert other effects as well: they increase peripheral glucose utilization by two mechanisms of action, stimulating hepatic gluconeogenesis, and increasing the number and sensitivity of insulin receptors[78]. Impairment of the effect on insulin secretion that occurs during chronic administration of sulfonylureas is due to the downregulation of the receptor of sulfonylureas on the surface of β-cells.

    Compared with sulfonylureas, glinides, including repaglinide, nateglinide, and mitiglinide, are rapid - acting insulin secretagogue that mimics early-phase insulin release, thereby providing better control of the postprandial glucose levels. For instance, repaglinide, which is known as an oral non-sulfonylurea insulin secretagogue, plays a powerful role in postprandial blood glucose reduction by promoting the release of postprandial insulin. It is widely used in patients with renal insufficiency for the reason that it is mainly metabolized in the liver by the cytochrome P450 (CYP) 3A4 enzyme system with a low risk of hypoglycemia[79]. Of note, studies have shown that repaglinide can be used in early treatment of cystic-fibrosis-related diabetes[80]. Compared with repaglinide, nateglinide, another glinide drug, has a faster onset and shorter action time. Additionally, it has been reported that glinides have long-term protective effects on β cell function and cardiovascular tissue[79]. Moreover, glinides have been widely used clinically and display excellent safety and efficacy and are associated with lower risk of hypoglycemia in comparison to sulfonylureas. Therefore, glinides are a good choice for monotherapy or combination therapy. However, the response to glinides varies among individuals, which is partially due to genetic factors involved in drug absorption, distribution, metabolism, targeting and different stages of disease.

    Mechanistically, glinides stimulate insulin secretion by inhibiting KATP in the pancreatic β-cell membrane. In addition to binding to SUR like sulfonylureas, repaglinide also binds to a unique site and blocks KATP in β-cells[81], which depolarizes the cell membrane to enhance calcium influx, eventually resulting in the release of insulin[82]. Repaglinide improves insulin sensitivity, and this effect is strengthened by inhibiting KCNQ1, the α-subunit of slow delayed rectifying potassium channel[83-84].

    It is known that hyperglycemia occurs only when insulin secretion is insufficient to overcome barriers to insulin action. Thus, when insulin action is normal, hyperglycemia occurs when absolute insulin secretion is deficient. In contrast, when insulin action is impaired (IR), hyperglycemia occurs when insulin secretion is inadequate to overcome IR. IR increases the prevalence of T2DM sixfold[85]. Thiazolidinediones, mainly including pioglitazone and rosiglitazone, are the pharmacologic agents that specifically treat IR. These drugs act as PPARγ agonists to increase insulin sensitivity and inhibit inflammation. Besides, thiazolidinedione can reduce fat deposition in liver, muscle, and pancreas, thus protecting them from the deleterious effects of free fatty acids. Importantly, thiazolidinedione provides long-term protection owing to the fact that progression of diabetes is delayed even if the drug is discontinued[86]. Astonishingly, studies have shown that rosiglitazone can continuously reduce fasting blood glucose for 3.5 years[87].

    The metabolic consequences of IR are increased risk of T2DM and associated cardiovascular diseases[85]. In this regard, the beneficial effects of thiazolidinediones on cardiovascular risk factors associated with IR have been well documented. In addition, thiazolidinediones also have protective effects on vascular endothelium[88]. Thiazolidinediones can reduce blood viscosity and platelet aggregation induced by IR. Furthermore, thiazolidinediones can reduce blood pressure, improve heart function, and promote fibrinolysis[89]. In addition, thiazolidinediones can also protect the kidney through a variety of mechanisms, including repressing inflammation and oxidative stress, reducing lipid deposition, improving renal microcirculation, and inhibiting the proliferation of glomerular and tubular cells, thus ultimately reducing the excretion of urinary protein, as well as preventing glomerulosclerosis and tubulointerstitial fibrosis[90-95]. Thiazolidinediones are now generic and less costly than pharmaceutical company-promoted therapies and are the most effective pharmacologic agents for treating IR.

    However, the use of thiazolidinediones has been limited because of the concern about the safety issues and side effects: there were increasing concerns about the thiazolidinediones-induced increases in fluid retention, risk of heart failure, weight gain, and peripheral fractures, and adverse effects on macular edema[96]. Thus, the use of these previously extensively prescribed and effective treatments for T2DM has been remarkably diminished because of concerns. To a certain extent, the above-mentioned renal protective effects of thiazolidinediones could also be ascribed to the inhibition of RAAS system[97].

    Alpha-glucosidase inhibitors (AGIs), including acarbose, miglitol, and voglibose, are recommended for therapy by the international Diabetes Federation (IDF)[98] and American Association of Clinical Endocrinologists (AACE)[99]. Their efficacy, safety, tolerability, cardiovascular benefits, and lack of hypoglycemia make them suitable for T2DM. They may also be used in patients with impaired glucose tolerance and can delay the occurrence of T2DM in these patients. They are particularly useful for patients who are at risk of hypoglycemia or lactic acidosis, and thus, are not suitable for other antidiabetic drugs such as sulfonylureas and metformin.

    Acarbose is a commonly used AGIs in clinic and it works by inhibiting upper gastrointestinal enzymes α-amylase and other α-glucosidases that break down complex carbohydrates into glucose, thus preventing absorption of starch and other carbohydrates from the brush border of the intestine and reducing postprandial glucose to improve glycemic control[100-101]. It also has an insulin-sparing effect and can increase incretin hormones and glucagon like peptide-1 but inhibit the postprandial release of gastric inhibitory polypeptide (GIP), helping reduce body weight[102]. Although acarbose may sometimes cause abdominal distension, it can effectively reduce weight and postprandial blood glucose, thus delaying the progression of prediabetes due to impaired glucose tolerance (IGT) to T2DM and reducing the risk of hypertension and CVD in IGT patients[103]. Moreover, acarbose can ameliorate vascular endothelial dysfunction and promote angiogenesis and wound healing, which may probably be mediated by the Akt/eNOS signaling pathway[104]. In addition, acarbose has been shown to prolong lifespan and enhance survival rate via decreasing postprandial blood glucose and insulin levels[105]. It is also reported that the intestinal microbiota play a significant role in life extension of acarbose[106]. Low-grade albuminuria, namely urinary albumin to creatinine ratio (UACR) < 30 mg/g, was reported to be associated with many cardiovascular risk factors and diabetic nephropathy[107-108]. Both acarbose and metformin can ameliorate low-grade albuminuria of T2DM[109].

    Overall, this class of compounds delays intestinal carbohydrate absorption, reduces postprandial glycaemia, and helps manage diabetes. Voglibose and miglitol inhibit the disaccharide-digesting enzymes but have no effect on the starch digesting enzyme α-amylase[110-111]. Of note, miglitol alleviates myocardial ischemia via inhibiting the decomposition of glycogen in myocardium, affecting ATP sensitive potassium channel, and activating GLP-1[112]. Miglitol may inhibit myocardial apoptosis by reducing the expression of Bax protein and the production of hydroxyl radical[113]. In addition, miglitol has inhibitory effect on α-glucosidase of human intestinal microflora[114]. AGIs can also exert beneficial effects on lipid metabolism and the treatment of acquired immune deficiency syndrome (AIDS) and tumors[115-117]. Collectively, AGIs are an effective, safe, and well tolerated treatment for diabetes, which provide cardiovascular benefits as well and can be used as monotherapy, combination therapy with other oral drugs and insulin, or fixed-dose combinations[118].

    DPP-Ⅳ inhibitors are ubiquitous enzymes that act on incretin hormones, mainly GLP-1 and gastric inhibitory peptide (GIP), which are critical for maintaining glucose homeostasis through increasing insulin secretion and decreasing glucagon secretion. By inhibiting the DPP-Ⅳ enzyme, DPP-Ⅳ inhibitors (alogliptin, sitagliptin, saxagliptin, linagliptin), also known as gliptins, increase the levels of GLP-1 and GIP, which in turn increase β-cell insulin secretion in pancreas, thereby reducing postprandial and fasting hyperglycemia[119]. On the other hand, glycemic variability is an emerging target for preventing complications related to T2DM, and DPP-Ⅳ inhibitors have shown their superior effectiveness in reducing glycemic variability compared to other oral anti-hyperglycemic drugs (OADs)[120]. A systematic review and meta-analysis of randomized controlled trials (RCTs) were performed to evaluate the effect of DPP-Ⅳ inhibitors compared with other anti-hyperglycemic drugs on glycemic variability[121].

    The findings indicate that DPP-Ⅳ inhibitors are promising alternatives for reducing glycemic variability in T2DM patients. Pharmacologically, DPP-Ⅳ inhibitors enhance GLP-1 preservation and expansion of β-cell mass through the inhibition of apoptotic pathways and improve blood glucose control without inducing hypoglycemia[122-123].

    In addition, vildagliptin has anti-inflammatory and lipid-improving effects[124]. Vildagliptin can protect against vascular diseases[125]. Furthermore, studies have shown that sitagliptin yields protective effects on the heart of mice and improves myocardial ischemia[126-127]. DPP-Ⅳ inhibitors can improve IR and inhibit liver inflammation and steatosis, therefore probably being used in the treatment of nonalcoholic fatty liver disease (NAFLD) in the future[128]. Sitagliptin was reported to have beneficial effects on intestinal flora[129]. DPP-Ⅳ inhibitors can protect vascular endothelium and prevent atherosclerosis, and the actions are independent of GLP-1 and GIP[130]. DPP-Ⅳ inhibitors might probably be used for the treatment of inflammatory bowel diseases (IBD) in the future [131]. Notably, DPP-Ⅳ inhibitors improve cardiac remodeling mainly through the following mechanisms: anti-inflammation, antioxidative stress and anti-cardiac fibrosis, thus exerting a preventive role in atrial fibrillation[132].

    Overall, DPP-Ⅳ inhibitors have good safety profiles and are worthy of wide application if the indications are appropriate. Although saxagliptin was associated with an increased rate of hospitalization for heart failure, DPP-Ⅳ inhibitors do not in general increase risk of cardiovascular death, non-fatal myocardial infarction or non-fatal stroke when compared to placebo in T2DM patients[133-134]. However, DPP-Ⅳ inhibitors can elicit some adverse effects. The most common side effects noticed in DPP-4 inhibitors sitagliptin and saxagliptin are upper respiratory tract infection, nasopharyngitis, headache, urinary tract infection, and arthralgia. They can also induce arthritis and arthralgia through various mechanisms such as cytokines release and inflammatory response[135]. It has been reported that DPP-Ⅳ may be negatively correlated with cancer, hence, long-term inhibition of DPP-Ⅳ enzyme in cancer may be detrimental[136]. However, some studies have also shown that DPP-Ⅳ inhibitors have anticancer properties due to cytotoxic effects[137].

    Sodium-glucose co-transporter 2 (SGLT2) inhibitors are the latest class of antidiabetic medications that work by inhibiting the absorption of glucose from the proximal tubule of the kidney and hence cause glycosuria, exerting a strong effect in reducing blood glucose. Four SGLT2 inhibitors are currently in clinical use, including canagliflozin, dapagliflozin, empagliflozin, and ertugliflozin. SGLT2 inhibitors reduce glycated hemoglobin by 0.5%-1.0%, improve glycemic control (fasting and postprandial), and reduce body weight[138]. Due to the virtue of no interference with insulin action and secretion, their efficacy remains the same even in presence of progressive β-cell dysfunction in T2DM. A meta-analysis unraveled that SGLT2 inhibitors achieved better glycemic control and greater weight reduction than DPP-Ⅳ inhibitors without increasing the risk of hypoglycemia in patients with T2DM that is inadequately controlled with insulin[139]. Recent clinical guidelines have suggested the use of SGLT2 inhibitors as add-on therapy in patients for whom metformin alone does not achieve glycemic goals, or as initial dual therapy with metformin in patients who present with higher glycated hemoglobin (HbA1c) levels[140]. Moreover, the addition of other beneficial actions on the top of improved glycemic control with a low risk of hypoglycemia with the combination of metformin and SGLT2 inhibitors is another attraction. However, caution needs to be taken of increased risk of euglycemic diabetic ketoacidosis during combination therapy.

    In addition to their hypoglycemic action, SGLT2 inhibitors can also produce protective effects on a variety of organs/systems. One of these beneficial actions is to reduce cardiovascular events, particularly heart failure, in cardiovascular outcome trials[141]. First, SGLT2 inhibitors have demonstrated impressive cardioprotective and reno-protective effects, and favorable effectiveness in reducing body weight, blood pressure, lipid profile, arterial stiffness, and endothelial function. Excessive sympathetic activity can result in increased arterial stiffness, endothelial dysfunction, and fluid retention[142-144]. SGLT2 inhibitors can alleviate sympathetic overactivity and reduce preload and afterload, thus lowering blood pressure without reflexive heart rate acceleration[145]. Studies have shown that empagliflozin improves endothelial function and delays the progression of aortic atherosclerotic plaque[146]. Of note, SGLT2 inhibitors can reduce the risk of heart failure and cardiac hypertrophy[147]. SGLT2 inhibitors may achieve cardiovascular protection through a variety of mechanisms, such as lowering blood pressure, correcting electrolyte disorders, alleviating mitochondrial dysfunction, improving energy metabolism, and suppressing cardiac fibrosis[148-149]. Second, SGLT2 inhibitors play a protective role in kidney by reducing urinary protein, constricting renal afferent arterioles, promoting uric acid excretion, inhibiting renal inflammation, attenuating the expression of fibrosis genes, and increasing autophagy, eventually postponing renal failure[150-154]. Besides, SGLT2 inhibitors can prevent podocyte loss, reduce renal lipid accumulation, thus delaying the progression of nephropathy[155]. SGLT2 inhibitors can reduce liver fat deposition and improve liver function with or without weight loss, thus yielding a certain therapeutic effect on NAFLD[156-158]. Chronic low-grade inflammation is a key factor for the development of T2DM and associated complications, especially atherosclerosis and coronary artery disease[159-161]. SGLT2 inhibitors can improve the function of adipose tissue through inhibiting the accumulation of M1-polarized macrophages and mitigating the levels of proinflammatory factors, such as adiponectin and leptin which can promote cardiac and renal fibrosis, thereby improving IR, attenuating inflammation and improving vascular endothelial function[162-165]. Additionally, dapagliflozin has been documented to reduce the activation of inflammatory bodies and regulate the phenotypic polarization of macrophages, thus resulting in ameliorating myocardial fibrosis[166-167]. Moreover, SGLT2 inhibitors can also attenuate inflammatory response of skeletal muscle and nervous system[168]. Furthermore, SGLT2 inhibitors are reported to have anti-cancer properties, for example, ipragliflozin suppresses the proliferation of breast cancer cells[169]. Nonetheless, SGLT2 inhibitors may increase the risk of stroke[170].

    The main adverse events of SGLT2 inhibitors include urinary tract and genital infections, as well as euglycemic diabetic ketoacidosis. Concerns have also been raised about the association of SGLT2 inhibitors with lower limb amputations, Fournier gangrene, risk of bone fractures, female breast cancer, male bladder cancer, orthostatic hypotension, and acute kidney injury[171]. SGLT2 inhibitors can occasionally cause skin reactions, such as systemic rashes and urticaria[172].

    GLP-1 is an endogenous incretin released from gut enteroendocrine cells, which yields many beneficial effects through binding to its receptor. GLP-1 can inhibit appetite, delay gastric emptying, inhibit the secretion of glucagon by islet α-cells, improve vascular endothelial function, lower blood pressure, and most importantly promote insulin secretion of islet β-cells[173-174]. These advantages make GLP-1 receptor an excellent target for T2DM management. GLP-1RAs are a class of drugs with a well-established efficacy and safety profile in patients with T2DM. Compounds belonging to GLP-1RAs are primarily derived from modifications based on either exendin-4 (a compound present in Gila monster venom) or human GLP-1 active fragment[175]. GLP-1RAs can be divided into two categories according to their differences in pharmacodynamics: short-term acting exenatide and lixisenatide and long-term acting albiglutide, dulaglutide, exenatide, liraglutide, and semaglutide. Short-term acting GLP-1RAs primarily delay gastric emptying (lowering postprandial glucose) and long-term acting agents affect both fasting glucose (via enhanced glucose-dependent insulin secretion and reduced glucagon secretion in the fasting state) and postprandial glucose (via enhanced postprandial insulin secretion and inhibition of glucagon secretion)[176]. Aside from their main mechanism of action, GLP-1RAs can also activate autophagy, e.g., exendin-4 reduces β-cell damage by improving autophagy clearance[177-179].

    In addition to their hypoglycemic efficacy, GLP-1RAs can reduce cardiac reperfusion injury, which may be helpful to the treatment of myocardial infarction[180]. In addition, exendin-4 improves cardiac remodeling and diastolic dysfunction[181], and liraglutide reduces the accumulation of fibroblasts and collagen deposition, thus improving cardiac fibrosis[182]. GLP-1RAs also produce neuroprotective and neurotrophic effects to improve the symptoms of Alzheimer's disease and Parkinson's disease[183-185]. Exendin-4, a GLP-1 analogue, can inhibit renal inflammation, reduce proteinuria, improve renal function and alleviate renal fibrosis[186]. Moreover, GLP-1RAs have a beneficial effect on blood lipids, which is manifested by the decrease of chylomicrons and triglycerides[187-188]. GLP-1RAs lessen hepatic fatty degeneration, inflammation, and hepatic fibrosis[189], as well as lung inflammation and fibrosis[190].

    GLP-1RAs have mild side effects of nausea, vomiting and hypersensitivity. Some studies have shown that GLP-1RAs are related to the occurrence of gallbladder diseases, such as gallstones and acute cholecystitis[191-193].

    Insulin remains a predominant life-saving medication for both T1DM and T2DM because insulin therapy is the most effective method of lowering blood glucose. Endogenous insulin secretion can restrict the fluctuation of the high surge of blood glucose levels, while imitating natural insulin with exogenous analogues remains a challenge as different insulin analogs (rapid acting, short acting, intermediate acting, and long acting) have different pharmacokinetic and pharmacodynamic properties and inconsistent effectiveness in overall hyperglycemia level and nocturnal hypoglycemia. Thus, whether the use of insulin analogues is appropriate is still a question in great scientific debate.

    A systemic review included 10 trials involving a total of 2 751 participants with 1 388 being randomized to receive insulin analogues and 1 363 to receive regular human insulin[194]. The duration of intervention ranged from 24 to 104 weeks, with a mean of about 41 weeks. The trial populations showed diversity in disease duration and inclusion and exclusion criteria. The analysis found no clear benefits of short-acting insulin analogues over regular human insulin in people with T2DM. Overall, the results from the 10 RCTs failed to provide solid evidence in favor of use of insulin analogues with insufficient data on patient-relevant outcomes, like all-cause mortality, microvascular or macrovascular complications and severe hypoglycemic episodes.

    Another meta-analysis compares the effects of long-term treatment with ultra-long-acting insulin analogues (insulin glargine U100 and U300, insulin detemir and insulin degludec) with NPH (neutral protamine Hagedorn) insulin (human isophane insulin) in adults with T2DM[195]. This analysis included 24 RCTs, of which 16 trials compared insulin glargine to NPH insulin, and eight trials compared insulin detemir to NPH insulin. In these trials, 3 419 patients with T2DM were randomized to insulin glargine and 1 321 to insulin detemir. The duration of the included trials ranged from 24 weeks to five years. The results show that while the effects on HbA1c were comparable, treatment with insulin glargine and insulin detemir resulted in fewer participants experiencing hypoglycemia when compared with NPH insulin. Treatment with insulin detemir also reduced the incidence of serious hypoglycemia. Approximately 1% patients treated with insulin detemir instead of NPH insulin benefited. It should be noted that in these trials, low blood glucose and HbA1c targets were set, which correspond to near normal or even non-diabetic blood glucose levels. Therefore, the authors concluded that the results from the trials are only applicable to people in whom such low blood glucose concentrations are targeted, which does not conform to the current guidelines recommending less-intensive blood glucose lowering for most patients with T2DM in daily practice (e.g., people with cardiovascular diseases, a long history of T2DM, who are susceptible to hypoglycemia or older people). Additionally, the low-certainty evidence and trial designs that did not meet the current clinical practice guidelines render it unclear whether consistent effects could be achieved in daily clinical practice.

    Although the clinical efficacy of regular human insulin has been well appreciated over 35 years, rapid-acting insulin analogues (insulin lispro, insulin aspart, and insulin glulisine) offer a couple of clinical advantages over regular human insulin. First, rapid-acting insulin analogues are engineered with a reduced tendency to aggregate as hexamers, which allows for rapid dissociation and absorption after a subcutaneous injection. Second, the more recently developed fast-acting insulin aspart (faster aspart) is an ultrafast-acting mealtime insulin that contains the conventional insulin aspart in a new formulation with the excipients niacinamide and L-arginine to achieve faster insulin absorption than regular human insulin and the conventional insulin aspart formulation[196].

    PTP1B is the main enzyme involved in insulin receptor desensitization by acting as a negative regulator of insulin and leptin signal transduction and is considered as a promising molecular target with its inhibition and a novel therapeutic strategy for the treatment of T2DM by enhancing insulin sensitivity in various cells[197]. Several PTP1B inhibitors have already been discovered, which act by interacting with the binding site of the enzyme surrounding the catalytic amino acid Cys215 and the neighboring area or with the allosteric site around Phe280[198]. PTP1B inhibitors improve the sensitivity of insulin receptors and have the potential to cure IR-related diseases. A large number of PTP1B inhibitors, either synthetic or isolated as bioactive agents from natural products, have developed and investigated for their ability to stimulate insulin signaling[198-202].

    PTP1B inhibitors enhance sensibility to insulin by restricting the activity of enzyme and have curing effects. These promising advantages stimulate enormous efforts in PTP1B drug discovery from natural products, synthetic heterocyclic scaffolds, or heterocyclic hybrid compounds. Various protocols are being formulated to boost the pharmacological effects of PTP1B inhibitors. Moreover, the new advancements suggest that it is possible to obtain small-molecule PTP1B inhibitors with the required potency and selectivity[203]. Furthermore, future endeavors via an integrated strategy of using medicinal chemistry and structural biology will hopefully result in potent and selective PTP1B inhibitors as well as safer and more effective oral drugs.

    At present, however, the structural homologies in the catalytic domain of PTP1B with other protein tyrosine phosphatases (PTPs) like leukocyte common antigen-related, CD45, SHP-2 and T-cell-PTP post a challenge to achieving selectivity. Thus, only highly selective molecules exhibiting desired effects with minimal side-effects may find their clinical application.

    Although careful control of glycemia and blood pressure can effectively stabilize or even decrease morbidity and mortality associated with diabetes, existing cardiovascular complications may or may not be effectively relieved. The polyol pathway is of prime importance in the pathogenesis of diabetic complications. Aldose reductase (AR) is an enzyme of aldoketo reductase superfamily that catalyzes the conversion of glucose to sorbitol in the polyol pathway of glucose metabolism, whose activation under hyperglycemic conditions leads to the development of chronic diabetic complication, and inflammatory and cytotoxic conditions, even under a normoglycemic state[204-205]. AR represents an excellent drug target, and in this context, ARIs have received much attention worldwide. Diminishing sorbitol flux through suppressing polyol pathway by ARIs is an emerging approach for the management of major complications of diabetes.

    Indeed, a huge effort has been made to disclose novel compounds able to inhibit AR. Several ARIs with the potential, supported by promising pre-clinical results, to address diabetic complications and inflammatory diseases are being developed lately. Natural compounds and plant extracts are the major resources of ARIs[205-206]. In addition, a variety of synthetic small molecules as well as natural compounds were reported to inhibit AR. The chemical structures of ARIs are diverse, although carboxylic acid derivatives are the most important and the largest class of ARIs due to the structural feature of carboxylate anion group which fits well in the active site of AR. ARIs developed to date vary structurally and can be grouped into the following subclasses accordingly: (1) acetic acid derivatives (epalrestat, alrestatin, zopalrestat, zenarestat, ponalrestat, lidorestat, and tolrestat), (2) cyclic amides (sorbinil, minalrestat, and fidarestat), and (3) phenolic derivatives (related to benzopyran-4-one and chalcone). Among these inhibitors, epalrestat is the only commercially available inhibitor till date in Japan and recently permitted for marketing in China and India for the treatment of diabetic complications[207-208]. In addition, some other ARIs such as sorbinil and ranirestat have been advanced into the late phase of clinical trials and found to be safe for human use. Several structurally diverse chemical classes including carboxylic acid derivatives and cyclic imides have been explored as ARIs and are being used in a number of disease conditions like inflammation, asthma, uveitis, cancers and diabetic complications as well[209-210].

    Apart from diabetic complications, an excess of AR is found in various human cancers, such as liver, colon, breast, and cervical cancer. ARIs have been reported as safe anti-inflammatory drugs. It is believed that use of ARIs could relieve some of the major health concerns such as diabetic complications, cancer, and cardiovascular diseases.

    G protein-coupled receptors (GPCRs) in the gut-brain-pancreas axis are key players in the postprandial control of metabolism and food intake. Human islets express nearly three hundred GPCRs, and many of them (more than 30 GPCRs) have been implicated in the development and progression of β-cell dysfunction, IR, obesity, and inflammation, which can lead to T2DM[211-213]. Because of the critical pathophysiological function of GPCRs in the context of diabetes, they have been be taken as new therapeutic targets for the treatment of T2DM[214-215]. Pharmaceutical companies are developing T2DM therapies that activate islet GPCRs, that is, GPCR agonists, including the incretin receptors (GLP1R, GIPR), GPR119, FFAR1 (GPR40), FFAR4 (GPR120) and the bile acid receptor GPBAR1 (TGR5)[215-217]. Yet, to date GLP-1 receptor is the only islet GPCR for which agonists are in current clinical use, as already described in a previous section. A number of small-molecule GPR119 agonists have been developed as new oral antidiabetic drugs, aiming to enhance insulin secretion and reduce appetite by targeting gut endocrine cells alongside pancreatic β-cells. Such agonists are thought to have a low risk of inducing hypoglycemia and a beneficial impact on body weight. Several GPR119 agonists have entered clinical studies; however, despite good evidence of efficacy in preclinical studies with animal models, they failed to yield metabolic benefits in humans with T2DM[218]. Hence, many of these compounds have failed either in phase Ⅰ or Ⅱ and none has progressed beyond phase Ⅱ[216].

    It was therefore proposed that highly desirable properties for new GPCR agonists would include: Gαs- or Gαq-coupled receptor activity on pancreatic β- and δ-cells, low receptor activity on α-cells, high activity on GLP-1-producing L cells and direct central nervous system effects to reduce food intake[215].

    We have witnessed a growing body of evidence from animal studies supporting the anti-diabetic properties of some dietary polyphenols as a dietary therapy for the prevention and management of T2DM. Commonly consumed dietary polyphenols include polyphenol-rich mixed diets, tea and coffee, chocolate and cocoa, cinnamon, grape, pomegranate, red wine, berries, and olive oil. Dietary polyphenols may inhibit α-amylase and α-glucosidase, glucose absorption in the intestine by sodium-dependent glucose transporter 1 (SGLT1), and hepatic glucose output and stimulate insulin secretion. Polyphenols may also enhance insulin-dependent glucose uptake, activate 5' AMPK, modify gut microbiome, and elicit anti-inflammatory effects. However, human epidemiological and intervention studies showed inconsistent results[219]. These facts prompted us to propose that discovery and development of single-compound natural products (NPs) with clear bioactivities might be the right way to go for exploring new natural anti-diabetic therapeutics. Indeed, NPs, particularly traditional herbal medicine and medicinal plants, have become an important resource of novel agents for drug discovery and have been extensively investigated for their anti-diabetic efficacy and potential clinical application to T2DM.

    The mechanisms of actions of the anti - diabetic NPs identified thus far are diverse, including inhibition of intestinal α-glucosidase and α-amylase, oxidative stress protection, formation of advanced glycation end products, aldose reductase, plasma glucose levels, enhancing enzyme activity of hexokinases and glucose-6-phosphate (G6P), postprandial hyperglycemia, the activity of G6P, and the level of skeletal hexokinases and increases in the synthesis and release of insulin and glucose transporter type 4 (GLUT4), etc.

    Kim et al.[219] and Hung et al. [220] provided an excellent and more detailed review on NPs and anti-diabetic therapy. Here we are just giving a few examples of well-known single-compound NPs for their possible applications to T2DM.

    Resveratrol is a non-flavonoid polyphenol that naturally occurs as phytoalexin, with the shell and stem of Vitis vinifera L (Vitaceae) as the richest source. Numerous experimental studies and clinical trials have documented the effectiveness and potential of resveratrol to benefit DM patients with reduction of blood glucose, improvement of insulin sensitivity and preservation of pancreatic β-cells, as well as alleviation of diabetic complications[221-222]. The therapeutic action and mechanisms of this compound on diabetes are complex and multifaceted. It has been speculated that use of resveratrol, alone or in combination with current anti-diabetic agents, might be a better approach for more effective management of DM and associated complications in the future[221, 223]. However, a recent meta-analysis on three RCTs with a total of 50 participants did not reach a conclusion as to whether resveratrol is beneficial or adverse following 4-5 weeks treatment at 10 mg to 1 000 mg T2DM patients due to inadequate evidence[224].

    Curcumin is a bioactive constituent present in the rhizome of the Curcuma longa plant, also known as turmeric. It has been reported to retard the development of diabetes, improve the function and prevent death of β-cells, and decrease IR[225]. The anti-diabetic activity of curcumin was also attributed to its potent suppressive effects on oxidative stress and inflammation. Moreover, it can also ameliorate the diabetes-induced endothelial dysfunction and downregulate the expression of nuclear factor-kappa B. Furthermore, curcumin has also been demonstrated to inhibit advanced glycation and collagen crosslinking[226]. A randomized, double-blinded, placebo-controlled trial including 240 prediabetic subjects showed that after curcumin (curcuminoids 250 mg/day) or placebo capsules for 9 months, none of subjects were diagnosed with diabetes, whereas 16.4% of the placebo group were diagnosed with T2DM[227]. A more recent study reported that curcumin supplementation (475 mg) for a period of 10 days attenuated hyperglycemia and hyperlipidemia in T2DM subjects treated with glyburide[228].

    Berberine, an isoquinoline alkaloid extracted from Coptis Root and Phellodendron Chinese, has been used as an adjuvant treatment of T2DM, hyperlipidemia, and hypertension in China. Animal and human studies have demonstrated this compound as a hypoglycemic agent in the management of diabetes[229-230]. A systemic review on 27 randomized controlled clinical trials with a total number of 2 569 patients revealed that berberine has comparable therapeutic effect on T2DM, hyperlipidemia and hypertension without serious side effect[231]. Another meta-analysis with 28 clinical studies of 2 313 T2DM patients focusing on fasting plasma glucose, postprandial plasma glucose and HbA1c levels suggests that the anti-diabetic efficacy of berberine combined with hypoglycemics is better than that with either berberine or hypoglycemics monotherapy[232]. Evidence has been presented that berberine lowers blood glucose in T2DM patients through increasing insulin receptor expression[233].

    In summary, T2DM is a heterogeneous and progressive systemic condition that represents a series of metabolic disorders characterized by hyperglycemia and caused by defects in insulin secretion and/or insulin action due to a complex network of pathological processes. It is driven by multiple genetic and environmental factors that interact and mutually reinforce one another leading to an increased risk of complications including heart, peripheral arterial and cerebrovascular disease, obesity, and nonalcoholic fatty liver disease, among others. At present, T2DM has become the third leading cause of human death. Studies have shown that the risk of death and cardiovascular events in diabetic patients is 1.5-2.0 times higher than that in healthy people[234-235].

    Although T2DM can be treated by a decent array of drugs, current pharmacologic therapy is not enough: straight glycemic control with neither cure of the disease per se nor effective prevention of associated complications. Even the control of blood sugar level is not as idea as we expect or as it should be, and sometimes the side effects elicited by the anti-diabetic drugs overwhelm their beneficial effects. Despite that intensive and extensive research has let identification of a large body of potential therapeutic targets for T2DM, agents with precise and specific targeting or alternatively broad-range targeting, on the key factors or pathways leading to the disease and its complications are presently lacking. For example, while it has been known that IR is a key element of T2DM, none of the market drugs targets the enhancement of the action of the intracellular part of insulin receptor or recuperation of the glucose transport mechanism in GLUT4-dependent cells. This situation can be partly ascribed to our insufficient understanding of etiopathogenesis and mechanisms of T2DM. Clearly, there is still a need to identify additional therapeutic agents for better management of the disease and its complications.

    While there is nothing wrong for aiming to achieve optimal glycemic control, because uncontrolled glucose levels result in T2DM progression and increase the risk of complications and mortality, such a strategy often increases the risk of hypoglycemia. Several antihyperglycemic agents have been developed over time, which provides us an opportunity for prescribing anti-T2DM agents based on suitability for the individual patient's characteristics. Yet our poor understanding of pharmacogenetics, the branch of genetics that investigates how our genome influences individual responses to drugs, therapeutic outcomes, and incidence of adverse effects, prevents us from taking benefits. According to a systemic review, there are 40 polymorphisms for each drug class among metformin, DPP-4 inhibitors/GLP1R agonists, thiazolidinediones, and sulfonylureas/meglitinides[236]. This finding suggests the possibility of genetic screening of variants/loci for identifying the precise therapeutic targets and for decision-making on the therapeutic approach through precision medicine. Such an approach is anticipated to improve the efficacy and safety of anti-diabetic pharmacotherapy of T2DM on one hand and to reduce the economic burden on a global scale on the other hand. Some therapies for diabetes increase the risk of hypoglycemia, in particular all insulins and insulin secretagogues, including the glinides and sulfonylureas. Hypoglycemia remains a major limiting factor to successful glycemic management, despite the availability of prevention options such as insulin analogues, continuous glucose monitoring, insulin pumps, and dogs that have been trained to detect hypoglycemia. It is often necessary to rely on combination therapy of multiple drugs or insulin. For example, combination treatment with a GLP-1 agonist and basal insulin has been proposed as a treatment strategy for T2DM that could provide robust glucose-lowering capability with low risk of hypoglycemia or weight gain. GLP-1 agonist and basal insulin combination treatment can enable achievement of the ideal trifecta in diabetic treatment: robust glycemic control with no increased hypoglycemia[237]. However, combination drug therapy bears some inherent challenges, e.g., different agents with different pharmacokinetics that are not easy to mutually compromise, possible drug interactions, difficulty of determining optimal dosages, etc. A superior approach that could avoid the weaknesses of combinational drug therapy is the one based on the idea of "one drug-multiple targets"[238-240] or polypharmacology[241], a new subdiscipline of pharmacology for the design or use of pharmaceutical agents that act on multiple targets or disease pathways. Whilst there have not been any rationally designed multitarget drugs for diabetes, "one drug-multiple targets" or polypharmacology would be a right direction for our future endeavors for the cure of T2DM and its complications.

    An exciting part of the findings from basic research is the identification and characterization of the primary pathophysiological factors as the potential therapeutic targets for T2DM and its complications in addition to deficient insulin secretion and IR[242]. Evolving findings unravel the critical roles of glucose transport, adipokine dysregulation, immune deregulation, chronic/low-grade inflammation, gut microbiota anomalies, and mitochondrial dysfunction in the pathogenesis and progression of T2DM and its associated complications. These findings lay the groundwork for future development of new agents that specifically and precisely act on one of the newly identified therapeutic targets or that integrate appropriate elements for multiple targeting based on the principle of polypharmacology.

    Acknowledgements: This work was supported by the National Natural Science Foundation of China (NSFC) (No. 81770809).

    Conflicts of interests

    All authors declare no competing interests.

  • [1]
    Benito-Vicente A, Jebari S, Larrea-Sebal A, et al. Pathophysiology of type 2 diabetes mellitus. Int J Mol Sci, 2020; 21(17): 6275. doi: 10.3390/ijms21176275
    [2]
    Chatterjee S, Khunti K, Davies M J. Type 2 diabetes. Lancet, 2017; 389(10085): 2239-2251. doi: 10.1016/S0140-6736(17)30058-2
    [3]
    Stumvoll M, Goldstein B J, van Haeften T W. Type 2 diabetes: principles of pathogenesis and therapy. Lancet, 2005; 365(9467): 1333-1346. doi: 10.1016/S0140-6736(05)61032-X
    [4]
    Yoon K H, Lee J H, Kim J W, et al. Epidemic obesity and type 2 diabetes in Asia. Lancet, 2006; 368(9548): 1681-1688. doi: 10.1016/S0140-6736(06)69703-1
    [5]
    Weyer C, Bogardus C, Mott D M, et al. The natural history of insulin secretory dysfunction and insulin resistance in the pathogenesis of type 2 diabetes mellitus. J Clin Invest, 1999; 104(6): 787-794. doi: 10.1172/JCI7231
    [6]
    Christensen A A, Gannon M. The beta cell in type 2 diabetes. Curr Diab Rep, 2019; 19(9): 81. doi: 10.1007/s11892-019-1196-4
    [7]
    Halban P A, Polonsky K S, Bowden D W, et al. Beta-cell failure in type 2 diabetes: postulated mechanisms and prospects for prevention and treatment. Diabetes Care, 37(6): 1751-1758.
    [8]
    Czech M P. Insulin action and resistance in obesity and type 2 diabetes. Nat Med, 2017; 23(7): 804-814. doi: 10.1038/nm.4350
    [9]
    Pearson T, Wattis J A, King J R, et al. The effects of insulin resistance on individual tissues: an application of a mathematical model of metabolism in humans. Bull Math Biol, 2016; 78(6): 1189-1217. doi: 10.1007/s11538-016-0181-1
    [10]
    Wilcox G. Insulin and insulin resistance. Clin Biochem Rev, 2005; 26(2): 19-39.
    [11]
    Cerf M E. Beta cell dysfunction and insulin resistance. Front Endocrinol (Lausanne), 2013; 4: 37.
    [12]
    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(2): 88-98. doi: 10.1038/nrendo.2017.151
    [13]
    Battisti W P, Palmisano J, Keane W E. Dyslipidemia in patients with type 2 diabetes. relationships between lipids, kidney disease and cardiovascular disease. Clin Chem Lab Med, 2003; 41(9): 1174-1181.
    [14]
    Verges B. Pathophysiology of diabetic dyslipidaemia: where are we? Diabetologia, 2015; 58(5): 886-899. doi: 10.1007/s00125-015-3525-8
    [15]
    Taskinen M R. Diabetic dyslipidaemia: from basic research to clinical practice. Diabetologia, 2003; 46(6): 733-749. doi: 10.1007/s00125-003-1111-y
    [16]
    Chapman M J. Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: evidence and guidance for management. Eur Heart J, 2011; 32(11): 1345-1361. doi: 10.1093/eurheartj/ehr112
    [17]
    Nordestgaard B G, Varbo A. Triglycerides and cardiovascular disease. Lancet, 2014; 384(9943): 626-635. doi: 10.1016/S0140-6736(14)61177-6
    [18]
    Yamamoto W R. Endoplasmic reticulum stress alters ryanodine receptor function in the murine pancreatic beta cell. J Biol Chem, 2019; 294(1): 168-181. doi: 10.1074/jbc.RA118.005683
    [19]
    Lynch S V, Pedersen O. The human intestinal microbiome in health and disease. N Engl J Med, 2016; 375(24): 2369-2379. doi: 10.1056/NEJMra1600266
    [20]
    Li X, Watanabe K, Kimura I. Gut microbiota dysbiosis drives and implies novel therapeutic strategies for diabetes mellitus and related metabolic diseases. Front Immunol, 2017; 8: 1882. doi: 10.3389/fimmu.2017.01882
    [21]
    Tang C, Ahmed K, Gille A, et al. Loss of FFA2 and FFA3 increases insulin secretion and improves glucose tolerance in type 2 diabetes. Nat Med, 2015; 21(2): 173-177. doi: 10.1038/nm.3779
    [22]
    Petersen K F, Befroy D, Dufour S, et al. Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science, 2003; 300(5622): 1140-1142. doi: 10.1126/science.1082889
    [23]
    Kim J A, Wei Y, Sowers J R. Role of mitochondrial dysfunction in insulin resistance. Circ Res, 2008; 102(4): 401-414. doi: 10.1161/CIRCRESAHA.107.165472
    [24]
    Schofield J H, Schafer Z T. Mitochondrial reactive oxygen species and mitophagy: a complex and nuanced relationship. Antioxid Redox Signal, 2021; 34(7): 517-530. doi: 10.1089/ars.2020.8058
    [25]
    Kadowaki T, Kadowaki H, Mori Y, et al. A subtype of diabetes mellitus associated with a mutation of mitochondrial DNA. N Engl J Med, 1994; 330(14): 962-968. doi: 10.1056/NEJM199404073301403
    [26]
    Tawata M, Hayashi J I, Isobe K, et al. A new mitochondrial DNA mutation at 14577 T/C is probably a major pathogenic mutation for maternally inherited type 2 diabetes. Diabetes, 2000; 49(7): 1269-1272. doi: 10.2337/diabetes.49.7.1269
    [27]
    Wang D, Taniyama M, Suzuki Y, et al. Association of the mitochondrial DNA 5178A/C polymorphism with maternal inheritance and onset of type 2 diabetes in Japanese patients. Exp Clin Endocrinol Diabetes, 2001; 109(7): 361-364.
    [28]
    Nesto R W. Correlation between cardiovascular disease and diabetes mellitus: current concepts. Am J Med, 2004; 116(Suppl 5A): 11S-22S.
    [29]
    Haffner S M, Lehto S, Ronnemaa T, et al. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med, 1998; 339(4): 229-234. doi: 10.1056/NEJM199807233390404
    [30]
    Beckman J A, Creager M A, Libby P. Diabetes and atherosclerosis: epidemiology, pathophysiology, and management. JAMA, 2002; 287(19): 2570-2581. doi: 10.1001/jama.287.19.2570
    [31]
    Holman R R, Paul S K, Bethel M A, et al. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med, 2008; 359(15): 1577-1589. doi: 10.1056/NEJMoa0806470
    [32]
    Turner R C, Cull C A, Frighi V, et al. Glycemic control with diet, sulfonylurea, metformin, or insulin in patients with type 2 diabetes mellitus: progressive requirement for multiple therapies (UKPDS 49). UK Prospective Diabetes Study (UKPDS) Group. JAMA, 1999; 281(21): 2005-2012.
    [33]
    Ihnat M A, Thorpe J E, Kamat C D, et al. Reactive oxygen species mediate a cellular 'memory' of high glucose stress signalling. Diabetologia, 2007; 50(7): 1523-1531. doi: 10.1007/s00125-007-0684-2
    [34]
    Olsen A S, Sarras Jr M P, Leontovich A, et al. Heritable transmission of diabetic metabolic memory in zebrafish correlates with DNA hypomethylation and aberrant gene expression. Diabetes, 2012; 61(2): 485-491. doi: 10.2337/db11-0588
    [35]
    Poy M N, Hausser J, Trajkovski M, et al. miR-375 maintains normal pancreatic alpha-and beta-cell mass. Proc Natl Acad Sci U S A, 2009; 106(14): 5813-5818. doi: 10.1073/pnas.0810550106
    [36]
    Reddy M A, Zhang E, Natarajan R. Epigenetic mechanisms in diabetic complications and metabolic memory. Diabetologia, 2015; 58(3): 443-455. doi: 10.1007/s00125-014-3462-y
    [37]
    Brasacchio D, Okabe J, Tikellis C, et al. Hyperglycemia induces a dynamic cooperativity of histone methylase and demethylase enzymes associated with gene-activating epigenetic marks that coexist on the lysine tail. Diabetes, 2009; 58(5): 1229-1236. doi: 10.2337/db08-1666
    [38]
    Al-Haddad R, Karnib N, Assaad R A, et al. Epigenetic changes in diabetes. Neurosci Lett, 2016; 625: 64-69. doi: 10.1016/j.neulet.2016.04.046
    [39]
    Ceriello A, Ihnat M A, Thorpe J E. Clinical review 2: The "metabolic memory": is more than just tight glucose control necessary to prevent diabetic complications? J Clin Endocrinol Metab, 2009; 94(2): 410-415. doi: 10.1210/jc.2008-1824
    [40]
    Nishikawa T, Edelstein D, Du X L, et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature, 2000; 404(6779): 787-790. doi: 10.1038/35008121
    [41]
    Reddy M A, Natarajan R. Epigenetic mechanisms in diabetic vascular complications. Cardiovasc Res, 2011; 90(3): 421-429. doi: 10.1093/cvr/cvr024
    [42]
    Guarner V, Rubio-Ruiz M E. Low-grade systemic inflammation connects aging, metabolic syndrome and cardiovascular disease. Interdiscip Top Gerontol, 2015; 40: 99-106.
    [43]
    Flory J, Lipska K. Metformin in 2019. JAMA, 2019; 322(13): 1926-1927.
    [44]
    American Diabetes Association. 9. Pharmacologic approaches to glycemic treatment: standards of medical care in diabetes-2019. Diabetes Care, 2019; 42(Supl 1): S90-S102.
    [45]
    Turner R C, Holman R R, Stratton I M. Effect of intensive bloodglucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet, 1998; 352(9131): 854-865. doi: 10.1016/S0140-6736(98)07037-8
    [46]
    De Vries S T, Denig P, Ekhart C, et al. Sex differences in adverse drug reactions of metformin: a longitudinal survey study. Drug Saf, 2020; 43(5): 489-495. doi: 10.1007/s40264-020-00913-8
    [47]
    Rena G, Hardie D G, Pearson E R. The mechanisms of action of metformin. Diabetologia, 2017; 60(9): 1577-1585. doi: 10.1007/s00125-017-4342-z
    [48]
    Knowler W C, Barrett-Connor E, Fowle S E, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med, 2002; 346(6): 393-403. doi: 10.1056/NEJMoa012512
    [49]
    Markowicz-Piasecka M, Sikora J, Szydlowska A, et al. Metformin-a future therapy for neurodegenerative diseases: theme: drug discovery, development and delivery in alzheimer's disease guest editor: davide brambilla. Pharm Res, 2017; 34(12): 2614-2627. doi: 10.1007/s11095-017-2199-y
    [50]
    Hundal R S, Inzucchi S E. Metformin: new understandings, new uses. Drugs, 2003; 63(18): 1879-1894. doi: 10.2165/00003495-200363180-00001
    [51]
    Martin-Montalvo A, Mercken E M, Mitchell S J, et al. Metformin improves healthspan and lifespan in mice. Nat Commun, 2013; 4: 2192. doi: 10.1038/ncomms3192
    [52]
    Pearce E L, Walsh M C, Cejas P J, et al. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature, 2009; 460(7251): 103-107. doi: 10.1038/nature08097
    [53]
    Sui X, Xu Y, Wang X, et al. Metformin: a novel but controversial drug in cancer prevention and treatment. Mol Pharm, 2015; 12(11): 3783-3791. doi: 10.1021/acs.molpharmaceut.5b00577
    [54]
    Podhorecka M, Ibanez B, Dmoszynska A. Metformin-its potential anti-cancer and anti-aging effects. Postepy Hig Med Dosw (Online), 2017; 71: 170-175.
    [55]
    McCreight L J, Bailey C J, Pearson E R. Metformin and the gastrointestinal tract. Diabetologia, 2016; 59(3): 426-435. doi: 10.1007/s00125-015-3844-9
    [56]
    Shaw R J, Lamia K A, Vasquez D, et al. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science, 2005; 310(5754): 1642-1646. doi: 10.1126/science.1120781
    [57]
    Singh A K, Gupta R, Ghosh A, et al. Diabetes in COVID-19: Prevalence, pathophysiology, prognosis and practical considerations. Diabetes Metab Syndr, 2020; 14(4): 303-310. doi: 10.1016/j.dsx.2020.04.004
    [58]
    Pal R, Bhadada S K. COVID-19 and diabetes mellitus: An unholy interaction of two pandemics. Diabetes Metab Syndr, 2020; 14(4): 513-517. doi: 10.1016/j.dsx.2020.04.049
    [59]
    Bornstein S R, Rubino F, Khunti K, et al. Practical recommendations for the management of diabetes in patients with COVID-19. Lancet Diabetes Endocrinol, 2020; 8(6): 546-550. doi: 10.1016/S2213-8587(20)30152-2
    [60]
    Scheen A J. Metformin and COVID-19: From cellular mechanisms to reduced mortality. Diabetes Metab, 2020; 46(6): 423-426. doi: 10.1016/j.diabet.2020.07.006
    [61]
    Huang I, Lim M A, Pranata, R. Diabetes mellitus is associated with increased mortality and severity of disease in COVID-19 pneumonia-A systematic review, meta-analysis, and meta-regression. Diabetes Metab Syndr, 2020; 14(4): 395-403. doi: 10.1016/j.dsx.2020.04.018
    [62]
    Bramante C T, Ingraham N E, Murray T A, et al. Observational Study of Metformin and Risk of Mortality in Patients Hospitalized with Covid-19. Lancet Healthy Longev, 2021; 2(1): e34-e41. doi: 10.1016/S2666-7568(20)30033-7
    [63]
    Sola D, Rossi L, Schianca G P C, et al. Sulfonylureas and their use in clinical practice. Arch Med Sci, 2015; 11(4): 840-848.
    [64]
    Melander A, Lebovitz H E, Faber O K. Sulfonylureas. Why, which, and how? Diabetes Care, 1990; 13(Suppl 3): 18-25.
    [65]
    Marshall A, Gingerich R L, Wright P H. Hepatic effect of sulfonylureas. Metabolism, 1970; 19(12): 1046-1052. doi: 10.1016/0026-0495(70)90028-4
    [66]
    Landstedt-Hallin L, Adamson U, Lins P E. Oral glibenclamide suppresses glucagon secretion during insulin-induced hypoglycemia in patients with type 2 diabetes. J Clin Endocrinol Metab, 1999; 84(9): 3140-3145.
    [67]
    Scarsi M, Podvinec M, Roth A, et al. Sulfonylureas and glinides exhibit peroxisome proliferator-activated receptor gamma activity: a combined virtual screening and biological assay approach. Mol Pharmacol, 2007; 71(2): 398-406. doi: 10.1124/mol.106.024596
    [68]
    Müller G. Dynamics of plasma membrane microdomains and crosstalk to the insulin signalling cascade. FEBS Lett, 2002; 531(1): 1-87. doi: 10.1016/S0014-5793(02)03452-X
    [69]
    Nakano N, Miyazawa N, Sakurai T, et al. Gliclazide inhibits proliferation but stimulates differentiation of white and brown adipocytes. J Biochem, 2007; 142(5): 639-645. doi: 10.1093/jb/mvm172
    [70]
    Sena C M, Louro T, Matafome P, et al. Antioxidant and vascular effects of gliclazide in type 2 diabetic rats fed high-fat diet. Physiol Res, 2009; 58(2): 203-209.
    [71]
    Lee K Y, Kim J R, Choi H C. Gliclazide, a KATP channel blocker, inhibits vascular smooth muscle cell proliferation through the CaMKKbeta-AMPK pathway. Vascul Pharmacol, 2018; 102: 21-28. doi: 10.1016/j.vph.2018.01.001
    [72]
    Rados D V, Pinto L C, Remonti L R, et al. Correction: The association between sulfonylurea use and all-cause and cardiovascular mortality: a meta-analysis with trial sequential analysis of randomized clinical trials. PLoS Med, 13(6): e1002091 doi: 10.1371/journal.pmed.1002091
    [73]
    Sehra D, Sehra S. Hypertension in type 2 diabetes mellitus: do we need to redefine the role of sulfonylureas? Recent Adv Cardiovasc Drug Discov, 2015; 10(1): 4-9.
    [74]
    Webb D R, Davies M J, Jarvis J, et al. The right place for Sulphonylureas today. Diabetes Res Clin Pract, 2019; 157: 107836. doi: 10.1016/j.diabres.2019.107836
    [75]
    Thisted H, Johnsen S P, Rungby J. Sulfonylureas and the risk of myocardial infarction. Metabolism, 2006; 55(5 Suppl 1): S16-S19.
    [76]
    Ramracheya R, Ward C, Shigeto M, et al. Membrane potentialdependent inactivation of voltage-gated ion channels in alpha-cells inhibits glucagon secretion from human islets. Diabetes, 2010; 59(9): 2198-2208. doi: 10.2337/db09-1505
    [77]
    Braun M, Ramracheya R, Amisten S, et al. Somatostatin release, electrical activity, membrane currents and exocytosis in human pancreatic delta cells. Diabetologia, 2009; 52(8): 1566-1578. doi: 10.1007/s00125-009-1382-z
    [78]
    Blumenthal S A. Potentiation of the hepatic action of insulin by chlorpropamide. Diabetes, 1977; 26(5): 485-489. doi: 10.2337/diab.26.5.485
    [79]
    Lv W, Wang X, Xu Q, et al. Mechanisms and characteristics of sulfonylureas and glinides. Curr Top Med Chem, 2020; 20(1): 37-56. doi: 10.2174/1568026620666191224141617
    [80]
    Ballmann M, Hubert D, Assael B M, et al. Repaglinide versus insulin for newly diagnosed diabetes in patients with cystic fibrosis: a multicentre, open-label, randomised trial. Lancet Diabetes Endocrinol, 2018; 6(2): 114-121. doi: 10.1016/S2213-8587(17)30400-X
    [81]
    Fuhlendorff J, Rorsman P, Kofod H, et al. Stimulation of insulin release by repaglinide and glibenclamide involves both common and distinct processes. Diabetes, 1998; 47(3): 345-351. doi: 10.2337/diabetes.47.3.345
    [82]
    Davies M J. Insulin secretagogues. Curr Med Res Opin, 2002; 18(Suppl 1): s22-s30.
    [83]
    Zhou X Y, Zhu J, Bao Z J, et al. A variation in KCNQ1 gene is associated with repaglinide efficacy on insulin resistance in Chinese Type 2 Diabetes Mellitus Patients. Sci Rep, 2016; 6: 37293. doi: 10.1038/srep37293
    [84]
    Xiao Z X, Chen R Q, Hu D X, et al. Identification of repaglinide as a therapeutic drug for glioblastoma multiforme. Biochem Biophys Res Commun, 2017; 488(1): 33-39. doi: 10.1016/j.bbrc.2017.04.157
    [85]
    Ford E S. Risks for all-cause mortality, cardiovascular disease, and diabetes associated with the metabolic syndrome: a summary of the evidence. Diabetes Care, 2005; 28(7): 1769-1778. doi: 10.2337/diacare.28.7.1769
    [86]
    Buchanan T A, Xiang A H, Peters R K, et al. Preservation of pancreatic beta-cell function and prevention of type 2 diabetes by pharmacological treatment of insulin resistance in high-risk hispanic women. Diabetes, 2002; 51(9): 2796-2803. doi: 10.2337/diabetes.51.9.2796
    [87]
    Diamant M, Heine R J. Thiazolidinediones in type 2 diabetes mellitus: current clinical evidence. Drugs, 2003; 63(13): 1373-1405. doi: 10.2165/00003495-200363130-00004
    [88]
    Reusch J E, Regensteiner J G, Watson P A. Novel actions of thiazolidinediones on vascular function and exercise capacity. Am J Med, 2003; 115(Suppl 8A): 69S-74S.
    [89]
    Giles T D, Sander G E. Effects of thiazolidinediones on blood pressure. Curr Hypertens Rep, 2007; 9(4): 332-337. doi: 10.1007/s11906-007-0060-0
    [90]
    Ko G J, Kang Y S, Han S Y, et al. Pioglitazone attenuates diabetic nephropathy through an anti-inflammatory mechanism in type 2 diabetic rats. Nephrol Dial Transplant, 2008; 23(9): 2750-2760. doi: 10.1093/ndt/gfn157
    [91]
    Sarafidis P A, Grekas D M. Insulin resistance and oxidant stress: an interrelation with deleterious renal consequences? J Cardiometab Syndr, 2007; 2(2): 39-142.
    [92]
    Pistrosch F, Herbrig K, Kindel B, et al. Rosiglitazone improves glomerular hyperfiltration, renal endothelial dysfunction, and microalbuminuria of incipient diabetic nephropathy in patients. Diabetes, 2005; 54(7): 2206-2211. doi: 10.2337/diabetes.54.7.2206
    [93]
    Sarafidis P A, Bakris G L. Protection of the kidney by thiazolidinediones: an assessment from bench to bedside. Kidney Int, 2006; 70(7): 1223-1233. doi: 10.1038/sj.ki.5001620
    [94]
    Buckingham R E, Al-barazanji K A, Toseland C D, et al. Peroxisome proliferator-activated receptor-gamma agonist, rosiglitazone, protects against nephropathy and pancreatic islet abnormalities in Zucker fatty rats. Diabetes, 1998; 47(8): 1326-1334.
    [95]
    Yoshimoto T, Naruse M, Nishikawa M, et al. Antihypertensive and vasculo-and renoprotective effects of pioglitazone in genetically obese diabetic rats. Am J Physiol, 1997; 272(6 Pt 1): E989-E996.
    [96]
    Nesto R W, Bell D, Bonow R O, et al. Thiazolidinedione use, fluid retention, and congestive heart failure: a consensus statement from the American Heart Association and American Diabetes Association. Diabetes Care, 2004; 27(1): 256-263. doi: 10.2337/diacare.27.1.256
    [97]
    Sarafidis P A, Stafylas P C, Georgianos P I, et al. Effect of thiazolidinediones on albuminuria and proteinuria in diabetes: a metaanalysis. Am J Kidney Dis, 2010; 55(5): 835-847. doi: 10.1053/j.ajkd.2009.1
    [98]
    Tseng Y H, Tsan Y T, Chan W C, et al. Use of an alpha-glucosidase inhibitor and the risk of colorectal cancer in patients with diabetes: a nationwide, population-based cohort study. Diabetes Care, 2015; 38(11): 2068-2074. doi: 10.2337/dc15-0563
    [99]
    Tamez-Perez H E, Proskauer-Pena S L, Hernrndez-Coria M I, et al. AACE comprehensive diabetes management algorithm 2013. Endocrine practice. Endocr Pract, 2013; 19(4): 736-737. doi: 10.4158/EP13210.LT
    [100]
    Van de Laa F A, Kucassen P L B, Akkermans R P, et al. Alphaglucosidase inhibitors for type 2 diabetes mellitus. Cochrane Database Syst Rev, 2005; 2005(2): CD003639.
    [101]
    Standl E, Schnell O, McGuire D K. Heart failure considerations of antihyperglycemic medications for type 2 diabetes. Circ Res, 2016; 118(11): 1830-1843. doi: 10.1161/CIRCRESAHA.116.306924
    [102]
    Hoffmann J, Spengler M. Efficacy of 24-week monotherapy with acarbose, glibenclamide, or placebo in NIDDM patients. The Essen Study. Diabetes Care, 1994; 17(6): 561-566. doi: 10.2337/diacare.17.6.561
    [103]
    Chiasson J L. Acarbose for the prevention of diabetes, hypertension, and cardiovascular disease in subjects with impaired glucose tolerance: the Study to Prevent Non-Insulin-Dependent Diabetes Mellitus (STOP-NIDDM) Trial. Endocr Pract, 2006; 12(Suppl 1): 25-30.
    [104]
    Han X, Deng Y P, Yu J W, et al. Acarbose accelerates wound healing via Akt/eNOS signaling in db/db Mice. Oxid Med Cell Longev, 2017; 2017: 7809581.
    [105]
    Dodds S G, Parihar M, Javors M, et al. Acarbose improved survival for Apc(+/Min) mice. Aging Cell, 2020; 19(2): e13088.
    [106]
    Smith B J, Miller R A, Ericsson A C, et al. Changes in the gut microbiome and fermentation products concurrent with enhanced longevity in acarbose-treated mice. BMC Microbiol, 2019; 19(1): 130. doi: 10.1186/s12866-019-1494-7
    [107]
    Kang S H, Jung D J, Choi E W, et al. Association between lowgrade albuminuria and hearing impairment in a non-diabetic Korean population: The Korea National Health and Nutrition Examination Survey (2011-2013). Ann Med, 2015; 47(8): 664-672. doi: 10.3109/07853890.2015.1093163
    [108]
    Chida S, Fujita Y, Ogawa A, et al. Levels of albuminuria and risk of developing macroalbuminuria in type 2 diabetes: historical cohort study. Sci Rep, 2016; 6: 26380. doi: 10.1038/srep26380
    [109]
    Pan Q, Xu Y, Yang N, et al. Metformin or acarbose treatment significantly reduced albuminuria in patients with newly diagnosed type 2 diabetes mellitus and low-grade albuminuria. Med Sci Monit, 2018; 24: 8941-8949. doi: 10.12659/MSM.911979
    [110]
    Chen X, Zheng Y, Shen Y. Voglibose (Basen, AO-128), one of the most important alpha-glucosidase inhibitors. Curr Med Chem, 2006; 13(1): 109-116. doi: 10.2174/092986706789803035
    [111]
    Scott L J, Spencer C M. Miglitol: a review of its therapeutic potential in type 2 diabetes mellitus. Drugs, 2000; 59(3): 521-549. doi: 10.2165/00003495-200059030-00012
    [112]
    Wang H, Shen Y, Zhao L, et al. 1-Deoxynojirimycin and its derivatives: a mini review of the literature. Curr Med Chem, 2021; 28(3): 628-643. doi: 10.2174/0929867327666200114112728
    [113]
    Wang N, Minatoguchi S, Chen X H, et al. Antidiabetic drug miglitol inhibits myocardial apoptosis involving decreased hydroxyl radical production and Bax expression in an ischaemia/reperfusion rabbit heart. Br J Pharmacol, 2004; 142(6): 983-990. doi: 10.1038/sj.bjp.0705863
    [114]
    Tan K, Tesar C, Wilton R, et al. Interaction of antidiabetic alphaglucosidase inhibitors and gut bacteria alpha-glucosidase. Protein Sci, 2018; 27(8): 1498-1508. doi: 10.1002/pro.3444
    [115]
    Kojima Y, Kimura T, Nakagawa K, et al. Effects of mulberry leaf extract rich in 1-deoxynojirimycin on blood lipid profiles in humans. J Clin Biochem Nutr, 2010; 47(2): 155-161. doi: 10.3164/jcbn.10-53
    [116]
    Silva C H, Taft C A. Computer-aided molecular design of novel glucosidase inhibitors for AIDS treatment. J Biomol Struct Dyn, 2004; 22(1): 59-63. doi: 10.1080/07391102.2004.10506981
    [117]
    Wang R J, Yang C H, Hu M L. 1-Deoxynojirimycin inhibits metastasis of B16F10 melanoma cells by attenuating the activity and expression of matrix metalloproteinases-2 and -9 and altering cell surface glycosylation. J Agric Food Chem, 2010; 58(16): 8988-8993. doi: 10.1021/jf101401b
    [118]
    American Diabetes Association. Standards of medical care in diabetes——2012. Diabetes Care 35(Suppl 1): S11-S63.
    [119]
    Capuano A, Sportiello L, Maiorino M I, et al. Dipeptidyl peptidase-4 inhibitors in type 2 diabetes therapy——focus on alogliptin. Drug Des Devel Ther, 2013; 7: 989-1001.
    [120]
    Rizzo M R, Barbieri M, Marfella R, et al. Response to comment on: Rizzo et al. Reduction of oxidative stress and inflammation by blunting daily acute glucose fluctuations in patients with type 2 diabetes: role of dipeptidyl peptidase-Ⅳ inhibition. Diabetes Care, 2012; 35: 2076-2082.
    [121]
    Lee S, Lee H, Kim Y, et al. Effect of DPP-Ⅳ inhibitors on glycemic variability in patients with T2DM: a systematic review and meta-analysis. Sci Rep, 2019; 9(1): 13296. doi: 10.1038/s41598-019-49803-9
    [122]
    Drucker D J, Nauck M A. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet, 2006; 368(9548): 1696-1705. doi: 10.1016/S0140-6736(06)69705-5
    [123]
    Drucker D J. The biology of incretin hormones. Cell Metab, 2006; 3(3): 153-165. doi: 10.1016/j.cmet.2006.01.004
    [124]
    Inaba W, Mizukami H, Kamata K, et al. Effects of long-term treatment with the dipeptidyl peptidase-4 inhibitor vildagliptin on islet endocrine cells in non-obese type 2 diabetic Goto-Kakizaki rats. Eur J Pharmacol, 2012; 691(1-3): 297-306. doi: 10.1016/j.ejphar.2012.07.030
    [125]
    Kröller-Schön S, Knorr M, Hausding M, et al. Glucose-independent improvement of vascular dysfunction in experimental sepsis by dipeptidyl-peptidase 4 inhibition. Cardiovasc Res, 2012; 96(1): 140-149. doi: 10.1093/cvr/cvs246
    [126]
    Ye Y M, Keyes K T, Zhang C F, et al. The myocardial infarct sizelimiting effect of sitagliptin is PKA-dependent, whereas the protective effect of pioglitazone is partially dependent on PKA. Am J Physiol Heart Circ Physiol, 2010; 298(5): H1454-H1465. doi: 10.1152/ajpheart.00867.2009
    [127]
    Read P A, Khan F Z, Heck P M, et al. DPP-4 inhibition by sitagliptin improves the myocardial response to dobutamine stress and mitigates stunning in a pilot study of patients with coronary artery disease. Circ Cardiovasc Imaging, 2010; 3(2): 195-201.
    [128]
    Yilmaz Y, Atug O, Yonal O, et al. Dipeptidyl peptidase Ⅳ inhibitors: therapeutic potential in nonalcoholic fatty liver disease. Med Sci Monit 2009; 15(4): HY1-5.
    [129]
    Liao X Y, Song L Y, Zeng B H, et al. Alteration of gut microbiota induced by DPP-4i treatment improves glucose homeostasis. EBioMedicine, 2019; 44: 665-674. doi: 10.1016/j.ebiom.2019.03.057
    [130]
    Shah Z, Kampfrath T, Deiuliis J A, et al. Long-term dipeptidylpeptidase 4 inhibition reduces atherosclerosis and inflammation via effects on monocyte recruitment and chemotaxis. Circulation, 2011; 124(21): 2338-2349. doi: 10.1161/CIRCULATIONAHA.111.041418
    [131]
    Salaga M, Binienda A, Draczkowski P, et al. Novel peptide inhibitor of dipeptidyl peptidase Ⅳ (Tyr-Pro-D-Ala-NH2) with anti-inflammatory activity in the mouse models of colitis. Peptides, 2018; 108: 34-45. doi: 10.1016/j.peptides.2018.08.011
    [132]
    Zhang X W, Zhang Z W, Li M Z, et al. Potential role of dipeptidyl peptidase-4 inhibitors in atrial fibrillation. Int J Cardiol, 2016; 207: 46-47.
    [133]
    Green J B, Bethel M A, Armstrong P W, et al. Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med, 2015; 373(3): 232-242. doi: 10.1056/NEJMoa1501352
    [134]
    Rosenstock J, Perkovic V, Johansen O E, et al. Effect of linagliptin vs placebo on major cardiovascular events in adults with type 2 diabetes and high cardiovascular and renal risk: the CARMELINA randomized clinical trial. JAMA, 2019; 321(1): 69-79. doi: 10.1001/jama.2018.18269
    [135]
    Mascolo A, Rafaniello C, Sportiello L, et al. Dipeptidyl peptidase (DPP)-4 inhibitor-induced arthritis/arthralgia: a review of clinical cases. Drug Saf, 2016; 39(5): 401-407. doi: 10.1007/s40264-016-0399-8
    [136]
    De S, Banerjee S, Kumar S K A, et al. Critical role of dipeptidyl peptidase Ⅳ: a therapeutic target for diabetes and cancer. Mini Rev Med Chem, 2019; 19(2): 88-97.
    [137]
    Amritha C A, Kumaravelu P, Chellathai D D. Evaluation of anti cancer effects of DPP-4 inhibitors in colon cancer-an invitro study. J Clin Diagn Res, 2015; 9(12): FC14-16.
    [138]
    Pandey J, Tamrakar A K. SGLT2 inhibitors for the treatment of diabetes: a patent review (2013-2018). Expert Opin Ther Pat, 2019; 29(5): 369-384. doi: 10.1080/13543776.2019.1612879
    [139]
    Min S H, Yoon J H, Hahn S, et al. Comparison between SGLT2 inhibitors and DPP4 inhibitors added to insulin therapy in type 2 diabetes: a systematic review with indirect comparison meta-analysis. Diabetes Metab Res Rev, 2017; 33(1): 27155214.
    [140]
    Sonne D P, Hemmingsen B. Comment on American diabetes association. Standards of medical care in diabetes-2017. Diabetes Care, 2017; 40(Suppl. 1): S1-S135. .
    [141]
    Verma S, McMurray J J V. SGLT2 inhibitors and mechanisms of cardiovascular benefit: a state-of-the-art review. Diabetologia, 2018; 61(10): 2108-2117. doi: 10.1007/s00125-018-4670-7
    [142]
    Mangoni A A, Mircoli L, Giannattasio C, et al. Effect of sympathectomy on mechanical properties of common carotid and femoral arteries. Hypertension, 1997; 30(5): 1085-1088. doi: 10.1161/01.HYP.30.5.1085
    [143]
    Hijmering M L, Stroes E S G, Olijhoek J, et al. Sympathetic activation markedly reduces endothelium-dependent, flow-mediated vasodilation. J Am Coll Cardiol, 2002; 39(4): 683-688. doi: 10.1016/S0735-1097(01)01786-7
    [144]
    DiBona G F. Sympathetic nervous system and the kidney in hypertension. Curr Opin Nephrol Hypertens, 2002; 11(2): 197-200. doi: 10.1097/00041552-200203000-00011
    [145]
    Lytvyn Y, Bjornstad P, Udell J A, et al. Sodium glucose cotransporter-2 inhibition in heart failure: potential mechanisms, clinical applications, and summary of clinical trials. Circulation, 2017; 136(17): 1643-1658. doi: 10.1161/CIRCULATIONAHA.117.030012
    [146]
    Han J H, Oh T J, Lee G, et al. The beneficial effects of empagliflozin, an SGLT2 inhibitor, on atherosclerosis in ApoE (-/-) mice fed a western diet. Diabetologia, 2017; 60(2): 364-376. doi: 10.1007/s00125-016-4158-2
    [147]
    Zelniker T A, Braunwald E. Cardiac and renal effects of sodiumglucose co-transporter 2 inhibitors in diabetes: JACC state-of-the-art review. J Am Coll Cardiol, 2018; 72(15): 1845-1855. doi: 10.1016/j.jacc.2018.06.040
    [148]
    Liu B, Wang Y, Zhang Y, et al. Mechanisms of protective effects of SGLT2 inhibitors in cardiovascular disease and renal dysfunction. Curr Top Med Chem, 2019; 19(20): 1818-1849. doi: 10.2174/1568026619666190828161409
    [149]
    Maejima Y. SGLT2 inhibitors play a salutary role in heart failure via modulation of the mitochondrial function. Front Cardiovasc Med, 2019; 6: 186.
    [150]
    Ishibashi Y, Matsui T, Yamagishi S. Tofogliflozin, a highly selective inhibitor of SGLT2 blocks proinflammatory and proapoptotic effects of glucose overload on proximal tubular cells partly by suppressing oxidative stress generation. Horm Metab Res, 2016; 48(3): 191-195.
    [151]
    Zinman B, Wanner C, Lachin J M, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med, 2015; 373(22): 2117-2128. doi: 10.1056/NEJMoa1504720
    [152]
    Kawanami D, Matoba K, Takeda Y, et al. SGLT2 Inhibitors as a therapeutic option for diabetic nephropathy. Int J Mol Sci, 2017; 18(5): 1083. doi: 10.3390/ijms18051083
    [153]
    Terami N, Ogawa D, Tachibana H, et al. Long-term treatment with the sodium glucose cotransporter 2 inhibitor, dapagliflozin, ameliorates glucose homeostasis and diabetic nephropathy in db/db mice. PLoS One 9, 2014; 9(6): e100777. doi: 10.1371/journal.pone.0100777
    [154]
    Lee Y H, Kim S H, Kang J M, et al. Empagliflozin attenuates diabetic tubulopathy by improving mitochondrial fragmentation and autophagy. Am J Physiol Renal Physiol, 2019; 317(4): F767-F780. doi: 10.1152/ajprenal.00565.2018
    [155]
    Wang X X, Levi J, Lu Y H, et al. SGLT2 protein expression is increased in human diabetic nephropathy: SGLT2 protein inhibition decreases renal lipid accumulation, inflammation, and the development of nephropathy in diabetic mice. J Biol Chem, 2017; 292(13): 5335-5348. doi: 10.1074/jbc.M117.779520
    [156]
    Shibuya T, Fushimi N, Kawai M, et al. Luseogliflozin improves liver fat deposition compared to metformin in type 2 diabetes patients with non-alcoholic fatty liver disease: A prospective randomized controlled pilot study. Diabetes Obes Metab, 2018; 20(2): 438-442. doi: 10.1111/dom.13061
    [157]
    Leiter L A, Forst T, Polidori D, et al. Effect of canagliflozin on liver function tests in patients with type 2 diabetes. Diabetes Metab, 2016; 42(1): 25-32. doi: 10.1016/j.diabet.2015.10.003
    [158]
    Komiya C, Tsuchiya K, Shiba K, et al. Ipragliflozin improves hepatic steatosis in obese mice and liver dysfunction in type 2 diabetic patients irrespective of body weight reduction. PLoS One, 2016; 11(3): e0151511. doi: 10.1371/journal.pone.0151511
    [159]
    Prattichizzo F, Nigris V D, Spiga R, et al. Inflammageing and metaflammation: The Yin and Yang of type 2 diabetes. Ageing Res Rev, 2018; 41: 1-17. doi: 10.1016/j.arr.2017.10.003
    [160]
    Libby P. Inflammation in atherosclerosis. Arterioscler Thromb Vasc Biol, 2012; 32(9): 2045-2051. doi: 10.1161/ATVBAHA.108.179705
    [161]
    Karasawa T, Takahashi M. Role of NLRP3 inflammasomes in atherosclerosis. J Atheroscler Thromb, 2017; 24(5): 443-451. doi: 10.5551/jat.RV17001
    [162]
    Garvey W T, Van Gaal L, Leiter L A, et al. Effects of canagliflozin versus glimepiride on adipokines and inflammatory biomarkers in type 2 diabetes. Metabolism, 2018; 85: 32-37. doi: 10.1016/j.metabol.2018.02.002
    [163]
    Xu L, Ota T. Emerging roles of SGLT2 inhibitors in obesity and insulin resistance: Focus on fat browning and macrophage polarization. Adipocyte, 2018; 7(2): 121-128.
    [164]
    Packer M. Do sodium-glucose co-transporter-2 inhibitors prevent heart failure with a preserved ejection fraction by counterbalancing the effects of leptin? A novel hypothesis. Diabetes Obes Metab, 2018; 20(6): 1361-1366. doi: 10.1111/dom.13229
    [165]
    Diaz-Rodríguez E, Agra R M, Fernández Á L, et al. Effects of dapagliflozin on human epicardial adipose tissue: modulation of insulin resistance, inflammatory chemokine production, and differentiation ability. Cardiovasc Res, 2018; 114(2): 336-346. doi: 10.1093/cvr/cvx186
    [166]
    Ye Y, Bajaj M, Yang H C, et al. SGLT-2 Inhibition with dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic cardiomyopathy in mice with type 2 diabetes. Further augmentation of the effects with saxagliptin, a DPP4 inhibitor. Cardiovasc Drugs Ther, 2017; 31(2): 119-132. doi: 10.1007/s10557-017-6725-2
    [167]
    Lee T M, Chang N C, Lin S Z. Dapagliflozin, a selective SGLT2 Inhibitor, attenuated cardiac fibrosis by regulating the macrophage polarization via STAT3 signaling in infarcted rat hearts. Free Radic Biol Med, 2017; 104: 298-310. doi: 10.1016/j.freeradbiomed.2017.01.035
    [168]
    Naznin F, Sakoda H, Okada T, et al. Canagliflozin, a sodium glucose cotransporter 2 inhibitor, attenuates obesity-induced inflammation in the nodose ganglion, hypothalamus, and skeletal muscle of mice. Eur J Pharmacol, 2017; 794: 37-44. doi: 10.1016/j.ejphar.2016.11.028
    [169]
    Komatsu S, Nomiyama T, Numata T, et al. SGLT2 inhibitor ipragliflozin attenuates breast cancer cell proliferation. Endocr J, 2020; 67(1): 99-106. doi: 10.1507/endocrj.EJ19-0428
    [170]
    Guo M, Ding J Y, Li J S, et al. SGLT2 inhibitors and risk of stroke in patients with type 2 diabetes: A systematic review and meta-analysis. Diabetes Obes Metab, 2018; 20(8): 1977-1982. doi: 10.1111/dom.13295
    [171]
    Tentolouris A, Vlachakis P, Tzeravini E, et al. SGLT2 inhibitors: a review of their antidiabetic and cardioprotective effects. Int J Environ Res Public Health, 2019; 16(16): 2965. doi: 10.3390/ijerph16162965
    [172]
    Yabe D, Nishikino R, Kaneko M, et al. Short-term impacts of sodium/glucose co-transporter 2 inhibitors in Japanese clinical practice: considerations for their appropriate use to avoid serious adverse events. Expert Opin Drug Saf, 2015; 14(6): 795-800. doi: 10.1517/14740338.2015.1034105
    [173]
    Lee Y S, Jun H S. Anti-inflammatory effects of GLP-1-based therapies beyond glucose control. Mediators Inflamm, 2016; 2016: 3094642.
    [174]
    Inzucchi S E, Bergenstal R M, Buse J B, et al. Management of hyperglycemia in type 2 diabetes, 2015: a patient-centered approach: update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care, 2015; 38(1): 140-149. doi: 10.2337/dc14-2441
    [175]
    Drucker D J. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab, 2018; 27(4): 740-756. doi: 10.1016/j.cmet.2018.03.001
    [176]
    Gentilella R, Pechtner V, Corcos A, et al. Glucagon-like peptide-1 receptor agonists in type 2 diabetes treatment: are they all the same? Diabetes Metab Res Rev, 2019; 35(1): e3070. doi: 10.1002/dmrr.3070
    [177]
    Arden C. A role for Glucagon-like peptide-1 in the regulation of beta-cell autophagy. Peptides, 2018; 100: 85-93. doi: 10.1016/j.peptides.2017.12.002
    [178]
    Zummo F P, Cullen K S, Honkanen-Scott M, et al. Glucagonlike peptide 1 protects pancreatic beta-cells from death by increasing autophagic flux and restoring lysosomal function. Diabetes, 2017; 66(5): 1272-1285. doi: 10.2337/db16-1009
    [179]
    Lim S W, Jin L, Jin J, et al. Effect of exendin-4 on autophagy clearance in beta cell of rats with tacrolimus-induced diabetes mellitus. Sci Rep, 2016; 6: 29921. doi: 10.1038/srep29921
    [180]
    Dokken B B, La Bonte L R, Davis-Gorman G, et al. Glucagon-like peptide-1 (GLP-1), immediately prior to reperfusion, decreases neutrophil activation and reduces myocardial infarct size in rodents. Horm Metab Res, 2011; 43(5): 300-305. doi: 10.1055/s-0031-1271777
    [181]
    Tate M, Robinson E, Green B D, et al. Exendin-4 attenuates adverse cardiac remodelling in streptozocin-induced diabetes via specific actions on infiltrating macrophages. Basic Res Cardiol, 2016; 111(1): 1. doi: 10.1007/s00395-015-0518-1
    [182]
    Gaspari T, Brdar M, Lee H W, et al. Molecular and cellular mechanisms of glucagon-like peptide-1 receptor agonist-mediated attenuation of cardiac fibrosis. Diab Vasc Dis Res, 2016; 13(1): 56-68. doi: 10.1177/1479164115605000
    [183]
    Heppner K M, Perez-Tilve D. GLP-1 based therapeutics: simultaneously combating T2DM and obesity. Front Neurosci, 2015; 9: 92.
    [184]
    Perry T, Lahiri D K, Sambamurti K, et al. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by abeta and iron. J Neurosci Res, 2003; 72(5): 603-612. doi: 10.1002/jnr.10611
    [185]
    de Graaf C, Donnelly D, Wootten D, et al. Glucagon-like peptide-1 and its class B G protein-coupled receptors: a long march to therapeutic successes. Pharmacol Rev, 2016; 68(4): 954-1013. doi: 10.1124/pr.115.011395
    [186]
    Kodera R, Shikata K, Kataoka H U, et al. Glucagon-like peptide-1 receptor agonist ameliorates renal injury through its anti-inflammatory action without lowering blood glucose level in a rat model of type 1 diabetes. Diabetologia, 2011; 54(4): 965-978. doi: 10.1007/s00125-010-2028-x
    [187]
    Hsieh J, Longuet C, Baker C L, et al. The glucagon-like peptide 1 receptor is essential for postprandial lipoprotein synthesis and secretion in hamsters and mice. Diabetologia, 2010; 53(3): 552-561. doi: 10.1007/s00125-009-1611-5
    [188]
    Xiao C, Bandsma R H, Dash S, et al. Exenatide, a glucagonlike peptide-1 receptor agonist, acutely inhibits intestinal lipoprotein production in healthy humans. Arterioscler Thromb Vasc Biol, 2012; 32(6): 1513-1519. doi: 10.1161/ATVBAHA.112.246207
    [189]
    Armstrong M J, Gaunt P, Aithal G P, et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet, 2016; 387(10019): 679-690. doi: 10.1016/S0140-6736(15)00803-X
    [190]
    Gou S, Zhu T, Wang W, et al. Glucagon like peptide-1 attenuates bleomycin-induced pulmonary fibrosis, involving the inactivation of NFkappaB in mice. Int Immunopharmacol, 2014; 22(2): 498-504. doi: 10.1016/j.intimp.2014.07.010
    [191]
    Faillie J L, Yu O H, Yin H, et al. Association of bile duct and gallbladder diseases with the use of incretin-based drugs in patients with type 2 diabetes mellitus. JAMA Intern Med, 2016; 176(10): 1474-1481. doi: 10.1001/jamainternmed.2016.1531
    [192]
    Holman R R, Bethel M A, Mentz R J, et al. Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med, 2017; 377(13): 1228-1239. doi: 10.1056/NEJMoa1612917
    [193]
    Marso S P, Daniels G H, Brown-Frandsen K, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med, 2016; 375(4): 311-322. doi: 10.1056/NEJMoa1603827
    [194]
    Fullerton B, Siebenhofer A, Jeitler K, et al. Short-acting insulin analogues versus regular human insulin for adult, non-pregnant persons with type 2 diabetes mellitus. Cochrane Database Syst Rev, 2018; 12(12): CD013228.
    [195]
    Semlitsch T, Engler J, Siebenhofer A, et al. (Ultra-)long-acting insulin analogues versus NPH insulin (human isophane insulin) for adults with type 2 diabetes mellitus. Cochrane Database Syst Rev, 2020; 11(11): CD005613.
    [196]
    Kruger D F, Novak L M. Role of ultrafast-acting insulin analogues in the management of diabetes. J Am Assoc Nurse Pract, 2019; 31(9): 537-548. doi: 10.1097/JXX.0000000000000261
    [197]
    Hussain H, Green I R, Abbas G, et al. Protein tyrosine phosphatase 1B (PTP1B) inhibitors as potential anti-diabetes agents: patent review (2015-2018). Expert Opin Ther Pat, 2019; 29(9): 689-702. doi: 10.1080/13543776.2019.1655542
    [198]
    Eleftheriou P, Geronikaki A, Petrou A. PTP1b inhibition, a promising approach for the treatment of diabetes type Ⅱ. Curr Top Med Chem, 2019; 19(4): 246-263. doi: 10.2174/1568026619666190201152153
    [199]
    Tamrakar A K, Maurya C K, Rai A K. PTP1B inhibitors for type 2 diabetes treatment: a patent review (2011-2014). Expert Opin Ther Pat, 2014; 24(10): 1101-1115. doi: 10.1517/13543776.2014.947268
    [200]
    Wang L J, Jiang B, Wu N, et al. Small molecules as potent protein tyrosine phosphatase 1B (PTP1B) inhibitors documented in patents from 2009 to 2013. Mini Rev Med Chem, 2015; 15: 104-122 doi: 10.2174/1389557515666150203144339
    [201]
    Verma M, Gupta S J, Chaudhary A, et al. Protein tyrosine phosphatase 1B inhibitors as antidiabetic agents-A brief review. Bioorg Chem, 2016; 70: 267-283.
    [202]
    Johnson T O, Ermolieff J, Jirousek M R. Protein tyrosine phosphatase 1B inhibitors for diabetes. Nat Rev Drug Discov, 2002; 1(9): 696-709. doi: 10.1038/nrd895
    [203]
    Duarte A M, Guarino M P, Barroso S, et al. Phytopharmacological strategies in the management of type 2 diabetes mellitus. Foods, 2020; 9(3): 271. doi: 10.3390/foods9030271
    [204]
    Grewal A S, Bhardwaj S, Pandita D, et al. Updates on aldose reductase inhibitors for management of diabetic complications and nondiabetic diseases. Mini Rev Med Chem, 2016; 16(2): 120-162.
    [205]
    Quattrini L, La Motta C. Aldose reductase inhibitors: 2013-present. Expert Opin Ther Pat, 2009; 29(3): 199-213.
    [206]
    Kawanishi K, Ueda H, Moriyasu M. Aldose reductase inhibitors from the nature. Curr Med Chem, 2003; 10(15): 1353-1374. doi: 10.2174/0929867033457304
    [207]
    Oka M, Kato N. Aldose reductase inhibitors. J Enzyme Inhib, 2001; 16(6): 465-473. doi: 10.1080/14756360127568
    [208]
    Ramunno A, Cosconati S, Sartini S, et al. Progresses in the pursuit of aldose reductase inhibitors: the structure-based lead optimization step. Eur J Med Chem, 2012; 51: 216-226. doi: 10.1016/j.ejmech.2012.02.045
    [209]
    Jedziniak J A, Kinoshita J H. Activators and inhibitors of lens aldose reductase. Invest Ophthalmol, 1971; 10(5): 357-366.
    [210]
    Kousaxidis A, Petrou A, Lavrentaki V, et al. Aldose reductase and protein tyrosine phosphatase 1B inhibitors as a promising therapeutic approach for diabetes mellitus. Eur J Med Chem, 2020; 207: 112742. doi: 10.1016/j.ejmech.2020.112742
    [211]
    Persaud S J. Islet G-protein coupled receptors: therapeutic potential for diabetes. Curr Opin Pharmacol, 2017; 37: 24-28. doi: 10.1016/j.coph.2017.08.001
    [212]
    Riddy D M, Delerive P, Summers R J, et al. G Protein-Coupled Receptors Targeting Insulin Resistance, Obesity, and Type 2 Diabetes Mellitus. Pharmacol Rev 2018; 70(1): 39-67. doi: 10.1124/pr.117.014373
    [213]
    Sebastiani G, Ceccarelli E, Castagna M G, et al. G-protein-coupled receptors (GPCRs) in the treatment of diabetes: Current view and future perspectives. Best Pract Res Clin Endocrinol Metab, 2018; 32(2): 201-213. doi: 10.1016/j.beem.2018.02.005
    [214]
    Hoque M, Ali S, Hoda M. Current status of G-protein coupled receptors as potential targets against type 2 diabetes mellitus. Int J Biol Macromol, 2018; 118(Pt 8): 2237-2244.
    [215]
    Reimann F, Gribble F M. G protein-coupled receptors as new therapeutic targets for type 2 diabetes. Diabetologia, 2006; 59(2): 229-233.
    [216]
    Ritter K, Buning C, Halland N, et al. G Protein-Coupled Receptor 119 (GPR119) agonists for the treatment of diabetes: recent progress and prevailing challenges. J Med Chem, 2016; 59(8): 3579-3592. doi: 10.1021/acs.jmedchem.5b01198
    [217]
    Mancini A D, Poitout V. GPR40 agonists for the treatment of type 2 diabetes: life after 'TAKing' a hit. Diabetes Obes Metab, 2015; 17(7): 622-629. doi: 10.1111/dom.12442
    [218]
    Katz L B, Gambale J J, Rothenberg P L, et al. Effects of JNJ-38431055, a novel GPR119 receptor agonist, in randomized, doubleblind, placebo-controlled studies in subjects with type 2 diabetes. Diabetes Obes Metab, 2012; 14(8): 709-716. doi: 10.1111/j.1463-1326.2012.01587.x
    [219]
    Kim Y, Keogh J B, Clifton P M. Polyphenols and glycemic control. Nutrients, 2016; 8(1): 17. doi: 10.3390/nu8010017
    [220]
    Hung H Y, Qian K, Morris-Natschke S L, et al. Recent discovery of plant-derived anti-diabetic natural products. Nat Prod Rep, 2012; 29(5): 580-606. doi: 10.1039/c2np00074a
    [221]
    Öztürk E, Arslan A K K, Yerer M B, et al. Resveratrol and diabetes: A critical review of clinical studies. Biomed Pharmacother, 2017; 95: 230-234. doi: 10.1016/j.biopha.2017.08.070
    [222]
    Szkudelski T, Szkudelska K. Resveratrol and diabetes: from animal to human studies. Biochim Biophys Acta, 2015; 1852(6): 1145-1154. doi: 10.1016/j.bbadis.2014.10.013
    [223]
    Timmers S, de Ligt M, Phielix E, et al. Resveratrol as add-on therapy in subjects with well-controlled type 2 diabetes: a randomized controlled trial. Diabetes Care, 2016; 39(12): 2211-2217. doi: 10.2337/dc16-0499
    [224]
    Jeyaraman M M, Al-Yousif N S H, Singh Mann A, et al. Resveratrol for adults with type 2 diabetes mellitus. Cochrane Database Syst Rev, 2020; 1(1): CD011919.
    [225]
    Pivari F, Mingione A, Brasacchio C, et al. Curcumin and type 2 diabetes mellitus: prevention and treatment. Nutrients, 2019; 11(8): 1837. doi: 10.3390/nu11081837
    [226]
    Nabavi S F, Thiagarajan R, Rastrelli L, et al. Curcumin: a natural product for diabetes and its complications. Curr Top Med Chem, 2015; 15(23): 2445-2455. doi: 10.2174/1568026615666150619142519
    [227]
    Chuengsamarn S, Rattanamongkolgul S, Luechapudiporn R, et al. Curcumin extract for prevention of type 2 diabetes. Diabetes Care, 2012; 35(11): 2121-2127. doi: 10.2337/dc12-0116
    [228]
    Neerati P, Devde R, Gangi A K. Evaluation of the effect of curcumin capsules on glyburide therapy in patients with type-2 diabetes mellitus. Phytother Res, 2014; 28(12): 1796-1800. doi: 10.1002/ptr.5201
    [229]
    Yin J, Xing H, Ye J. Efficacy of berberine in patients with type 2 diabetes mellitus. Metabolism, 2008; 57(5): 712-717. doi: 10.1016/j.metabol.2008.01.013
    [230]
    Lan J, Zhao Y Y, Dong F X, et al. Meta-analysis of the effect and safety of berberine in the treatment of type 2 diabetes mellitus, hyperlipemia and hypertension. J Ethnopharmacol, 2015; 161: 69-81. doi: 10.1016/j.jep.2014.09.049
    [231]
    Liang Y, Xu X J, Yin M J, et al. Effects of berberine on blood glucose in patients with type 2 diabetes mellitus: a systematic literature review and a meta-analysis. Endocr J, 2019; 66(1): 51-63.
    [232]
    Zhang H, Wei J, Xue R, et al. Berberine lowers blood glucose in type 2 diabetes mellitus patients through increasing insulin receptor expression. Metabolism, 2010; 59(2): 285-292. doi: 10.1016/j.metabol.2009.07.029
    [233]
    Emerging Risk Factors Collaboration, Di Angelantonio E, Kaptoge S, et al. Association of Cardiometabolic Multimorbidity With Mortality. JAMA, 2015; 314(1): 52-60. doi: 10.1001/jama.2015.7008
    [234]
    Dinesh Shah A, Langenberg C, Rapsomaniki E, et al. Type 2 diabetes and incidence of a wide range of cardiovascular diseases: a cohort study in 1.9 million people. Lancet, 2015; 385(Suppl 1): S86.
    [235]
    Mannino G C, Andreozzi F, Sesti G. Pharmacogenetics of type 2 diabetes mellitus, the route toward tailored medicine. Diabetes Metab Res Rev, 2019; 35(3): e3109. doi: 10.1002/dmrr.3109
    [236]
    Eng C, Kramer C K, Zinman B, et al. Glucagon-like peptide-1 receptor agonist and basal insulin combination treatment for the management of type 2 diabetes: a systematic review and meta-analysis. Lancet, 2014; 384(9961): 2228-2234. doi: 10.1016/S0140-6736(14)61335-0
    [237]
    Gao H, Xiao J N, Sun Q, et al. A single decoy oligodeoxynucleotides targeting multiple oncoproteins produces strong anticancer effects. Mol Pharmacol, 2006; 70(5): 1621-1629. doi: 10.1124/mol.106.024273
    [238]
    Lu Y, Xiao J N, Lin H X, et al. A single anti-microRNA antisense oligodeoxyribonucleotide (AMO) targeting multiple microRNAs offers an improved approach for microRNA interference. Nucleic Acids Res, 2009; 37(3): e24. doi: 10.1093/nar/gkn1053
    [239]
    Wang Z G. The concept of multiple-target anti-miRNA antisense oligonucleotide technology. Methods Mol Biol, 2011; 676: 51-57.
    [240]
    Roth B L, Sheffler D J, Kroeze W K. Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophrenia. Nat Rev Drug Discov, 2004; 3(4): 353-359. doi: 10.1038/nrd1346
    [241]
    Koutsouleris N, Meisenzahl EM, Borgwardt S, et al. Individualized differential diagnosis of schizophrenia and mood disorders using neuroanatomical biomarkers. Brain, 2015; 138(Pt 7): 2059-2073.
    [242]
    Schwartz S S, Epstein S, Corkey B E, et al. The time is right for a new classification system for diabetes: rationale and implications of the betacell-centric classification schema. Diabetes Care, 2016; 39(2): 179-186. doi: 10.2337/dc15-1585
  • Relative Articles

    [1]Lina Xuan, Huishan Luo, Shu Wang, Guangze Wang, Xingmei Yang, Jun Chen, Jianjun Guo, Xiaomeng Duan, Xiufang Li, Hua Yang, Shengjie Wang, Hailong Zhang, Qingqing Zhang, Shulei Liu, Yongtao She, Kai Kang, Lihua Sun. Circulating CCRR serves as potential novel biomarker for predicting acute myocardial infarction[J]. Frigid Zone Medicine, 2024, 4(3): 137-151. doi: 10.1515/fzm-2024-0015
    [2]Tongzhu Jin, Zhen Ye, Ruonan Fang, Yue Li, Wei Su, Qianqian Wang, Tianyu Li, Hongli Shan, Yanjie Lu, Haihai Liang. Notum protects against myocardial infarction-induced heart dysfunction by alleviating cardiac fibrosis[J]. Frigid Zone Medicine, 2024, 4(1): 41-50. doi: 10.2478/fzm-2024-0005
    [3]Donghua Liu, Yang Zhang, Xin Liu, Qihe Huang, Xiaofang Zhang, Rui Yang, Yue Zhao, Penghui Li, Jiayi He, Kexiao Zhang, Zhenwei Pan, Huiwen Liu, Baofeng Yang. GDF11 restores the impaired function of EPCs-MA by promoting autophagy: GDF11 ameliorates endothelial progenitor cell aging by promoting autophagy[J]. Frigid Zone Medicine, 2024, 4(4): 212-223. doi: 10.1515/fzm-2024-0021
    [4]Nannan Tang, Jiatong Li, Zhuo Wang, Jinlu Zuo, Zifeng Zhang, Di Huang, Yannan Han, Yuqing Chen, Yilin Sun, Xiang Li, Ruxue Mu, Qingxue Ma, Jie Zhang, Jiaying Wu, He Wang, Hongxia Zhao, Xingli Dong, Zhiguo Wang, Yu Liu, Dan Zhao, Baofeng Yang. Small ubiquitin-like modifiers inhibitors lower blood pressure via ERK5/KLF2-dependent upregulation of the eNOS/NO pathway[J]. Frigid Zone Medicine, 2024, 4(4): 202-211. doi: 10.1515/fzm-2024-0020
    [5]Wen Li, Jia Wang, Yilian Yang, Chunlei Duan, Bing Shao, Mingxiu Zhang, Jiapan Wang, Peifeng Li, Ye Yuan, Yan Zhang, Hongyu Ji, Xingda Li, Zhimin Du. Ethanol extract of cassia seed alleviates metabolic dysfunction-associated steatotic liver disease by acting on multiple lipid metabolism-related pathways[J]. Frigid Zone Medicine, 2024, 4(3): 160-176. doi: 10.1515/fzm-2024-0017
    [6]Liangliang Jiang, Qiushuang Qiao, Jing Wang. PLA2G2D and CHIT1: Potential biomarkers for immune infiltration and prognosis in cervical squamous cell carcinoma[J]. Frigid Zone Medicine, 2024, 4(2): 110-119. doi: 10.1515/fzm-2024-0012
    [7]Xianqi Sun, Yuzhen Li, Huiwen Yu, Jiaying Lin, Chen Wang, Quanlin Liu, Bingxue Bai. ISG15 promotes M5-induced hacat cell proliferation through Wnt signaling in psoriasis[J]. Frigid Zone Medicine, 2024, 4(4): 224-232. doi: 10.1515/fzm-2024-0022
    [8]Siyu Hao, Yu Zhang, Anqi Yin, Ying Lyu, Nannan Tong, Jiangtian Tian, Yuzhen Li. Toll-like receptors 2 polymorphism is associated with psoriasis: A case-control study in the northern Chinese population[J]. Frigid Zone Medicine, 2024, 4(2): 96-101. doi: 10.1515/fzm-2024-0010
    [9]Jiayue Zhang, Liyao Sun, Xiaohan Yu, Chen Yang, Qi An, Chaoqun Wei, Hongyan Ge. LCN2 aggravates diabetic cataracts by promoting ferroptosis in lens epithelial cells[J]. Frigid Zone Medicine, 2024, 4(3): 177-192. doi: 10.1515/fzm-2024-0018
    [10]Han Lou, Henghui Xu, Yong Zhang. FAM210A: Implications in mitochondrial dynamics and metabolic health[J]. Frigid Zone Medicine, 2023, 3(4): 196-198. doi: 10.2478/fzm-2023-0025
    [11]Manyu Gong, Xuewen Yang, Yaqi Wang, Yanying Wang, Dongping Liu, Haodong Li, Yunmeng Qu, Xiyang Zhang, Yanwei Zhang, Han Sun, Lei Jiao, Ying Zhang. Growth differentiation factor 11 promotes macrophage polarization towards M2 to attenuate myocardial infarction via inhibiting Notch1 signaling pathway[J]. Frigid Zone Medicine, 2023, 3(1): 53-64. doi: 10.2478/fzm-2023-0008
    [12]Xiaowei Wan, Qiuhai Qin, Ruitang Xie, Xin Li, Min Su. Network pharmacology-based approach for exploring the biotargets and mechanisms of vitamin A for the treatment of diabetic foot ulcers[J]. Frigid Zone Medicine, 2023, 3(3): 186-192. doi: 10.2478/fzm-2023-0023
    [13]Yanan Ni, Dan Liu, Xiaona Zhang, Hong Qiao. Association of point in range with β-cell function and insulin sensitivity of type 2 diabetes mellitus in cold areas[J]. Frigid Zone Medicine, 2023, 3(4): 242-252. doi: 10.2478/fzm-2023-0031
    [14]Haijian Sun, Xiaowei Nie, Kangying Yu, Jinsong Bian. Therapeutic potential of gasotransmitters for cold stress-related cardiovascular disease[J]. Frigid Zone Medicine, 2022, 2(1): 10-24. doi: 10.2478/fzm-2022-0002
    [15]Baofeng Yang. Inaugural Editorial[J]. Frigid Zone Medicine, 2021, 1(1): 1-1. doi: 10.2478/fzm-2021-0001
    [16]Xinyi Liu, Yonghui Pan, Jingjing Wang, Lei Zhang, Junli Zhang. Changes of calcitonin gene-related peptide and other serological indicators in vestibular migraine patients[J]. Frigid Zone Medicine, 2021, 1(2): 111-118. doi: 10.2478/fzm-2021-0014
    [17]Yan Feng, Ying Li, Xinling Yang, Limin Han, Luning Wang, Shan Gao, Ruixue Yin, Xue Wang, Jiayang Li, Meiming Liu, Baiyan Li. Direct evidence of VEGF-mediated neuroregulation and afferent explanation of blood pressure dysregulation during angiogenic therapy[J]. Frigid Zone Medicine, 2021, 1(2): 119-126. doi: 10.2478/fzm-2021-0015
    [18]Xin Xing, Shiqiang Wang. Mammalian hibernation: a unique model for medical research[J]. Frigid Zone Medicine, 2021, 1(2): 65-68. doi: 10.2478/fzm-2021-0008
    [19]De-an Guo, Wenlong Wei, Changliang Yao, Jianqing Zhang, Qirui Bi. Chinese herbal medicines with beneficial effects[J]. Frigid Zone Medicine, 2021, 1(2): 79-83. doi: 10.2478/fzm-2021-0010
    [20]Yanyan Liu, Yahan Yu, Xinyao Wang, Guanqun Liu, Xinda Yin, Yunlong Bai, Zhimin Du. Overexpression of microRNA-135b-5p attenuates acute myocardial infarction injury through its antioxidant and anti-apptotic properties[J]. Frigid Zone Medicine, 2021, 1(2): 85-94. doi: 10.2478/fzm-2021-0011
  • Created with Highcharts 5.0.7Amount of accessChart context menuAbstract Views, HTML Views, PDF Downloads StatisticsAbstract ViewsHTML ViewsPDF Downloads2024-062024-072024-082024-092024-102024-112024-122025-012025-022025-032025-042025-0502.557.51012.515
    Created with Highcharts 5.0.7Chart context menuAccess Class DistributionFULLTEXT: 30.5 %FULLTEXT: 30.5 %META: 66.2 %META: 66.2 %PDF: 3.2 %PDF: 3.2 %FULLTEXTMETAPDF
    Created with Highcharts 5.0.7Chart context menuAccess Area Distributionother: 5.2 %other: 5.2 %China: 35.1 %China: 35.1 %Germany: 2.6 %Germany: 2.6 %Reserved: 1.9 %Reserved: 1.9 %United States: 55.2 %United States: 55.2 %otherChinaGermanyReservedUnited States

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)  / Tables(2)

    Article Metrics

    Article views (101) PDF downloads(5) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return