Next Article in Journal
Therapeutic Opportunities of Disrupting Genome Integrity in Adult Diffuse Glioma
Next Article in Special Issue
Influence of Type 2 Diabetes in the Association of PNPLA3 rs738409 and TM6SF2 rs58542926 Polymorphisms in NASH Advanced Liver Fibrosis
Previous Article in Journal
Gel-Forming of Self-Assembling Peptides Functionalized with Food Bioactive Motifs Modulate DPP-IV and ACE Inhibitory Activity in Human Intestinal Caco-2 Cells
Previous Article in Special Issue
Fatty Acid Synthase Inhibitor Platensimycin Intervenes the Development of Nonalcoholic Fatty Liver Disease in a Mouse Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring New Drug Targets for Type 2 Diabetes: Success, Challenges and Opportunities

1
Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bathinda 151001, India
2
University Institute of Pharmaceutical Sciences, UGC Centre for Advanced Studies, Panjab University, Chandigarh 160014, India
3
Department of Nuclear Medicine, Manipal College of Health Professions (MCHP), Manipal Academy of Higher Education (MAHE) Manipal, Manipal 576104, India
4
Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, India
5
Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
6
Department of Biomedical Engineering, Central University of Rajasthan, Ajmer 305817, India
*
Authors to whom correspondence should be addressed.
Biomedicines 2022, 10(2), 331; https://doi.org/10.3390/biomedicines10020331
Submission received: 11 January 2022 / Revised: 24 January 2022 / Accepted: 26 January 2022 / Published: 31 January 2022
(This article belongs to the Special Issue Metabolic Syndrome and NASH: From Molecular Basis to Therapy)

Abstract

:
There are substantial shortcomings in the drugs currently available for treatment of type 2 diabetes mellitus. The global diabetic crisis has not abated despite the introduction of new types of drugs and targets. Persistent unaddressed patient needs remain a significant factor in the quest for new leads in routine studies. Drug discovery methods in this area have followed developments in the market, contributing to a recent rise in the number of molecules. Nevertheless, troubling developments and fresh challenges are still evident. Recently, metformin, the most widely used first-line drug for diabetes, was found to contain a carcinogenic contaminant known as N-nitroso dimethylamine (NDMA). Therefore, purity and toxicity are also a big challenge for drug discovery and development. Moreover, newer drug classes against SGLT-2 illustrate both progress and difficulties. The same was true previously in the case of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Furthermore, researchers must study the importance of mechanistic characteristics of novel compounds, as well as exposure-related hazardous aspects of current and newly identified protein targets, in order to identify new pharmacological molecules with improved selectivity and specificity.

1. Epidemiology

Diabetes mellitus (DM) is a chronic progressive metabolic disorder in which the body is unable to utilize glucose [1]. It could be caused by a decrease in pancreatic cell insulin secretion or a lack of insulin responsiveness in the body. Insulin aids glucose assimilation within cells, allowing blood glucose levels to remain within a healthy range (80–120 mg/dL). Thus, a deficit of insulin in the body results in hyperglycemia, raising the blood glucose level, which in turn leads to many metabolic and life-threatening complications, including cardiovascular, nephropathic, and neuropathic diseases, amongst others [1].
A significant portion of the world’s population is affected by diabetes mellitus (DM). Studies estimate that 8.8% of those between the ages of 20–79 in the world’s population are diabetic [2]. According to an International Diabetes Federation (IDF) report [2], the Federated States of Micronesia (Micronesia) have the world’s most overweight population, with a 30% obesity prevalence [3]. On the other hand, North American and Caribbean adults aged between 20–79 years have the highest prevalence (13%) of diabetes among Western IDF regions [2]. In South Asian countries, diabetes prevalence is at its highest in Mauritius (22%), followed by Sri Lanka (10.7%), and India (10.4%) [2]. Approximately 425 million people worldwide are affected by diabetes, and this is projected to increase to ~629 million by the year 2045 due to unhealthy diets and inactive lifestyles [4].
In the case of Micronesia and other nearby islands, there is one hypothesis that argues the cause of severe diabetes is inadequate physical activity and an unhealthy diet. During times in which there is an inadequate food supply, people in Micronesia consume an excess amount of sugar to sustain themselves. After recovering from such periods, even with an increased availability of higher quality foods, sugar consumption has become an integral part of their daily dietary habits. Micronesian survey data show that around 73.1% of the population is overweight, and 32.1% frankly diabetic [5].
DM is a major threat to both the developed and developing worlds. According to the 2017 National Diabetes Statistics Report, about 30.3 million people in the U.S. are diabetic and surprisingly some 23.8% are undiagnosed [2]. The latest 2019 statistics by the IDF are shown in Table 1. However, in countries like India, the dominant factor contributing to the prevalence of diabetes is an underdeveloped health services system, as 70% of the Indian population lives in poorly serviced rural areas. Widespread illiteracy and unfamiliarity with healthier nutritional habits only serve to exacerbate the situation. These factors contribute to inadequate diabetes screening and availability of preventive measures, as well as non-adherence to diabetes management guidelines once diagnosed [6]. Less physical activity and sedentary lifestyles may also contribute to obesity and T2DM. Correspondingly, increased physical activity has been shown to help maintain glucose homeostasis in the body and to delay the onset of impaired glucose tolerance [7].

2. Classification of Diabetes Mellitus

Diabetes existed long before its mechanisms were understood. The first reference to diabetes was in 1550 B.C. in an Egyptian papyrus, where it was described as a rare disease characterized by excessive weight loss and frequent urination [8]. The term diabetes (to siphon or to pass through, in Greek) was first used by Apollonius of Memphis, who noted that patients with diabetes present with frequent urination [9]. A Greek physician, Galen, hypothesized it to be a type of kidney disease, as the most prominent symptom was glucosuria. Avicenna (980–1037) offered a comprehensive description of diabetes mellitus in his publication “The Canon of Medicine” in 1025 [10]. Eventually, owing to its characteristic symptoms, many tests to more fully identify the disease were developed in the late 11th century, involving the examination of the subject’s urine’s color, odor and taste. The sweet taste of a diabetic patient’s urine apparently led to its second name, mellitus (honey, in Latin) [9]. Later in the 19th century, Matthew Dobson discovered that the excessive amount of sugar in the urine of patients with diabetes gives it that sweet taste. He also provided an important distinction between the two different types of diabetes, which are now referred to as type 1 and type 2 diabetes. Banting and Macleod observed a reduction in blood glucose levels after administering islets of Langerhans to a pancreatectomized dog, and for this breakthrough discovery they were awarded the Nobel Prize in 1923 [11]. Eli Lilly acquired the first deal to produce insulin on a commercial level. Gradually, by the late 20th century, as the mechanisms behind diabetes started to unfold, various insulin preparations and other oral hypoglycemic drugs paved their way onto the market.
At present, based on insulin deficiency and insensitivity of cells towards insulin, diabetes is classified into three major types, described below. The detailed classification can be found in the WHO’s classification of diabetes mellitus 2019 [12].

2.1. Type 1 Diabetes Mellitus

T1DM is primarily caused by the destruction of pancreatic β-cells by the body’s immune system and is also known as Insulin Dependent Diabetes Mellitus. This type of diabetes is found across all age groups of patients, but the prevalence is higher among children [13].

2.2. Type 2 Diabetes Mellitus

T2DM is the most common type of diabetes. It is a chronic and multifactorial disease. Earlier this was also called Non-insulin Dependent Diabetes Mellitus (NIDDM), but this term is no longer used because insulin is often used in the management of NIDDM. T2DM is believed to be caused by the gradual development of cells resistant towards insulin and β-cell dysfunction, although the exact mechanism remains unclear. Being overweight or obese is a major risk factor for T2DM, as it may increase the likelihood of insulin resistance. This in turn will decrease uptake of glucose in, for example, the heart or musculoskeletal tissues, with simultaneous increase in glucose production in organs such as the liver [14]. To counter this, insulin secretion is enhanced by β-cells. Therefore, hyperglycemia and hyperinsulinemia often coexist during early T2DM [15]. In later stages of the disease, β-cell function is also diminished. Thus, therapeutics that target β-cell functioning and decrease insulin resistance are key treatments in T2DM. Some of the factors that control β-cell functioning include oxidative stress, ER-stress and autophagy. Over the years, researchers have worked on various targets to moderate the onset or worsening of diabetes in their patients, out of which many turned into drug targets.

2.3. Gestational Diabetes Mellitus (GDM)

Mothers with GDM is a type of glucose intolerance that occurs during pregnancy. It affects around 2–14% of pregnant women worldwide [16]. Mothers with GDM are at increased risk for the development of T2DM after pregnancy. Their offspring have increased rates of birth trauma, as well as a higher chance of developing issues with obesity and diabetes later in life. Mothers with GDM can be managed by diet and exercise with only some cases requiring medication.

3. Common Targets for Anti-Diabetic Drug

T1DM is managed by regular administration of insulin and modifications in the diet and lifestyle of affected individuals [17]. T2DM is the most common type of diabetes, and due to its complex pathogenesis, the ideal treatment for T2DM has been difficult to find. Several studies have reported compounds that improved insulin’s action on target tissues and helped restore β-cell function. In the last decade, many targets have been discovered as new oral agents for T2DM patients [18]. Among these, four major target types are known as insulin secretagogues, insulin mimickers and sensitizers, and starch blockers.

3.1. Insulin Secretagogues

Insulin secretagogues work by stimulating β-cells to secrete more insulin. There are two types: sulfonylureas and non-sulfonylureas. Sulphonylureas (SU) bind to the sulphonylurea receptors on pancreatic β-cells and stimulate them to secrete insulin. SU have shown good efficacy in reducing glycated hemoglobin microvasculature complications in patients with T2DM. However, their major drawback is prolonged binding time to the β-cells, which results in prolonged insulin release. The non-sulfonylureas also stimulate β-cells to secrete insulin, but they are short acting. In 2004, Jean-Claude Henquin proposed five potential action sites for insulin secretagogue β-cell metabolism activation: increased blocking of β-cell KATP channels; increase in intracellular (Ca2+) concentration by means other than KATP channels; activation of various amplifying pathways in β-cells; action on β-cell membrane receptors; and action on β-cell nuclear receptors [19].

3.2. Insulin Mimickers and Sensitizers

Insulin mimickers and sensitizers are agents which are helpful in lowering blood-glucose levels and are usually found as dietary supplements. They work by activating glucose transporters on muscle and fat cells, thus mimicking the function of insulin. Insulin sensitizers work by increasing the sensitivity of body tissues towards insulin. A variety of observable cardiac risk factors such as an increased risk of blood clotting, elevated blood pressure, lipid profile or C-reactive protein, undesired lipoprotein(a) or serum fibrinogen, and even abnormal thickening of the heart muscle have also been shown to improve upon use of insulin sensitizers [20].

3.3. Starch Blockers

Another important class is alpha-glucosidase inhibitors, which act by slowing down carbohydrate absorption. The most common drugs are Acarbose and miglitol [21]. Another category is dipeptidyl peptidase-4 (DPP-4) inhibitors, which also act in carbohydrate metabolism. For example, Glucagon-like peptide-1 (GLP-1) is released after meal ingestion and found highly expressed in adipose tissue. It works by delaying gastric emptying, increasing insulin secretion, and reducing glucagon secretion [22].
Sodium-glucose co-transporter type 2 (SGLT2) inhibitors are another recently discovered class. Canagliflozin and dapagliflozin are recently approved drugs in this category. These agents promote glucose excretion by the kidney and thus exert their actions independent of insulin (Figure 1a,b).
The other advantages of these drugs include modest weight loss and a low risk of hyperglycemia, etc. [23]. The search for the ideal anti-diabetic drug is still ongoing, leading to the development of newer molecules which are in different phases of clinical trials. Some of the anti-diabetic drugs at phase-III clinical trials are listed in Table 2.

4. Emerging Targets for the Treatment of T2DM

4.1. 11β-Hydroxysteroid Dehydrogenase

11β-Hydroxysteroid dehydrogenase is an enzyme that catalyzes the conversion of inactive cortisone to active cortisol. Literature reports indicated that high circulating levels of active glucocorticoid cortisol contribute to various disorders such as diabetes mellitus, obesity, dyslipidemia and high blood pressure. The 11β-HSD knockout transgenic mice were shown to have higher insulin sensitivity and were comparatively more resistant to high fat induced obesity, whereas studies also suggested that the over expression of 11β-HSD made animals prone to metabolic syndrome [24]. Hence, 11β-HSD is considered an important drug target for T2DM.

4.2. Glutamine Fructose-6-Phosphate Amido Transferase

As reported earlier, most glucose entering the cell undergo glycolytic metabolism and a very small percentage enter the hexosamine pathway. Biosynthesis of hexosamine is well known for its contribution to insulin resistance and activation of growth factor synthesis. Enzyme Glutamine fructose-6-phosphate aminotransferase (GFAT) has a central role in the hexosamine biosynthetic pathway, as it is involved in catalysis of the first and rate limiting steps for hexosamine formation. Therefore, it is considered to be an important therapeutic target against T2DM [25].

4.3. Protein Tyrosine Phosphatase 1B

Protein tyrosine phosphatases, such as protein tyrosine phosphatase 1B (PTP1B) and leukocyte antigen-related tyrosine phosphatase, play an important role in modulating insulin signal transductions [26]. Tyrosine phosphorylation in the insulin-receptor activation loop is an important step in insulin signal transduction. PTP1B dephosphorylates phosphor-tyrosine residues of insulin receptor kinase activation segments leading to negative regulation of insulin signaling. The evidence further suggests a relationship between PTP1B, insulin sensitivity, obesity and T2DM. PTP1B is also a negative regulator for leptin signaling [1]. Leptin is a hormone primarily involved in maintaining metabolic homeostasis, and in obesity a decreased sensitivity to leptin results in the inability to attain satiety despite having good energy stores. PTP1B also plays an important part in pancreatic β-cell proliferation. For example, Fernandez-Ruiz et al. found that that PTP1B knockout mice have a higher proliferation of β-cells and elevated glucose-induced insulin secretion [27]. These studies are strongly suggestive of the role of PTP1B in diabetes, and thus interest for PTP1B inhibitors, resulting in the search for and development of several inhibitors of this protein. Detailed descriptions of the reported PTP1B inhibitors can be found elsewhere [28].

4.4. SLC16A11

The genome-based studies carried out by the Slim Initiative in Genomic Medicine for the Americas (SIGMA) Type 2 Diabetes identified a genetic locus which is strongly associated with T2DM. These diabetes-associated haplotypes caused a decrease in SLC16A11 expression in the liver and disrupted interaction with basigin, which reduced the cell surface localization of SLC16A11 [29]. SLC16A11 knockdown in primary human hepatocytes leads to modulation of fatty acid and lipid metabolism [29]. This modulation increases intracellular acylcarnitine, diacylglycerol and triacylglycerol levels which results in increased triglycerides in blood circulation and an accumulation in liver tissue [30,31]. The increased levels of triglycerides are implicated in insulin resistance, so variants of SLC16A11 may increase the risk of diabetes by regulating lipid metabolism. However, many questions remain unanswered, for example, what the unique substrates are that are associated with the transport of mediators targeting the physiological and biochemical mechanisms that affect T2DM.

4.5. Nephroblastoma Overexpressed (CCN3/NOV)

CCN3 (also nephroblastoma overexpressed) is a cysteine-rich protein which is involved in growth-regulatory functions. Its presence is noted in a variety of human tissues and biological fluids such as the musculoskeletal system, kidneys, and cerebrospinal fluid [32]. The average plasma levels of CCN3 have a strong correlation with hs-CRP, BMI, and fat mass, and are elevated significantly in obese patients with hyperlipidemia [33]. Martinerie et al. demonstrated that CCN3 deficiency in mice fed with standard high-fat diets markedly lowered their body weight, and improved glucose tolerance and insulin sensitivity [34]. Moreover, Li et al. investigated the serum CCN3 levels in newly diagnosed T2DM (nT2DM) patients and compared it with healthy control subjects [35]. It was observed that CCN3 levels were significantly elevated in T2DM patients. These data suggest CCN3 may be involved in obesity-associated insulin-resistance and can serve as an important target for a T2DM therapeutic [35].

4.6. FoxO1

The forehead transcription factor FoxO1 is another important target of T2DM and a prominent mediator of insulin signaling in β-cells. It has been shown that dominant-negative FoxO1 adipocytes improved both glucose as well as insulin tolerance in a high-fat diet model. In addition, Fox01 in the pancreas are responsible for β-cell dysfunction by inducing stress and apoptosis. Phosphorylation and acetylation are the two most common post translational modifications of FoxO1, where it regulates many gene functions [36]. Phosphorylation of FoxO1 is the main mechanism by which it comes out of the nucleus to get degraded by ubiquitination. The acetylation state of FoxO1 depends upon the balance between the protein acetylases and deacetylases. Even FoxO1 gets activated by O-GlcNAcylation during oxidative stress-like conditions in diabetic livers and increases the activation of many gluconeogenic and ROS detoxifying genes [36]. PGC1-α induction during fasting activates the series of gluconeogenesis genes and later directly binds to FoxO1 to phosphorylate and ultimately degrade it out of the nucleus.

4.7. FFA2/FFA3

The free fatty acids (FFAs) can act as signaling molecules. FFAs are generally divided into three sub-categories based on their chain length as short-chain fatty acids (SCFAs), medium-chain fatty acids (MCFAs) and long-chain fatty acids (LCFAs). FFAs of these varying chain lengths are known to activate transmembrane receptors such as FFA1, FFA2, and FFA3. For example, FFA1 are expressed to a large extent on the pancreatic β-cells and are activated by LCFAs. The activation of these receptors also plays an important role in the increase of glucose-stimulated insulin secretion. Therefore, because of its obvious role in glucose-stimulated insulin secretion, a series of FFA1 ligands have been identified and tested. FFA2 and FFA3 are akin to FFA1 receptors and get activated by SCFAs but their role in insulin is complex. FFA2 receptors are highly expressed in immune cells, especially neutrophils [37,38,39]. Studies have also identified FFA2 expression on murine pancreatic β-cell line MIN6 and isolated mouse islets [40,41]. The indirect support for the role of FFA2 in diabetes comes from the fact that a fiber-rich diet is linked with a lower incidence of diabetes and SCFAs. Ligands for FFA2 are known to be produced by intestinal bacteria in the body by the fermentation of dietary fibers [42]. Tang et al. [43] showed FFA2 and FFA3 expression on human pancreatic β-cells. These receptors are shown to inhibit insulin secretion by coupling to Gi-type G proteins. They also demonstrated that genetic deletion of FFA2 and FFA3 receptors in pancreatic β-cells leads to higher insulin secretion in HFD animals, whereas similar deletion in intestinal cells did not affect glucose tolerance in diabetic animals. This study demonstrated that FFA2/FFA3 antagonists may prove beneficial in T2DM [43].

4.8. Epoxyeicosatrienoic Acids (EETs)

Epoxyeicosatrienoic acids (EETs) are produced from arachidonic acid by cytochrome p450 enzymes (monoxygenase/epoxygenase) in vascular endothelium responses to various stimuli, such as the agonists acetylcholine (ACH) or bradykinin, or by shear stress which activates phospholipase A2 to release arachidonic acid [9]. The cytochrome p450 enzyme is responsible for the formation of 20-hydroxyeicosatetraenoic acid (20-HETE) and EETs. The soluble epoxide hydrolases and reactive oxygen species (ROS) induction rapidly hydrolyze the EETs into their respective dihydroxyepoxytrienoic acids (DHETs) as well as esterificate primarily to glycerophospholipids [44]. The literature shows that EETs demonstrate anti-inflammatory, vasodilatory, and anti-apoptotic actions, inhibited sEH significantly, and elevated EETs’ cellular and circulation levels [45,46,47].
The administration of EET or inhibitors of sEH to obese mice is associated with a decrease in visceral subcutaneous fat and an increase in insulin sensitivity. The CYP2J2-mediated increase in EET increases fatty acid oxidation and adiposity. Although EETs have a naturally robust action, including vasodilatation and restraint of the inflammatory response, in adiposity, their effect on mitochondrial work and proliferator-actuated receptor gamma coactivator-1α (PGC-1α) corresponding to adipogenesis is still unclear [46,48,49,50].

4.9. Peroxisome Proliferator-Activated Receptor Gamma Co-Activator Alpha (PGC-1α)

PGC-1α is a protein encoded by the PPARGC1A gene in humans. PGC-1α maintains energy homeostasis and regulates the expression of insulin signaling, mitochondrial biogenesis, dynamics, and antioxidant genes, including uncoupling proteins, and thus prevents mitochondrial dysfunction and metabolic disorders related to adipocyte malfunction [51]. Dysregulation of PGC-1α alters homeostasis in cells and exacerbates the inflammatory response, which is commonly accompanied by metabolic disturbances. During adipocyte dysfunction, low degrees of PGC-1α downregulate mitochondrial quality articulation, instigate irritation and oxidative pressure, and advance atomic factor κ-B actuation [52].
PGC-1α quality treatment facilitated improved fat tissue work and had a positive and beneficial effect on distal organs like the liver, suggesting that focusing on PGC-1α quality treatment is an alluring remedial methodology for improving insulin affectability, insulin processing, metabolic movement and vascular capacity in metabolic disorders [53].

4.10. Peroxisome Proliferator-Activated Receptor Gama (PPAR©)

One of the most important nuclear targets for TZDs is PPAR©. Originally, it was considered the positive regulator of adipogenesis with higher expression in adipose tissues, though it is also expressed in many other tissues and body cells of the body but to a lesser extent. It has been observed that specific deletion of PPAR© from different cell targets—such as adipose tissue, muscle, macrophages, and the brain—alters glucose homeostasis, which in turn reduces the activity of TZDs. PPAR© is well known for both promoting and secreting adiponectin which is considered an important insulin sensitizing hormone [36]. It is a well-known transcription factor that regulates the functions of many genes both positively and negatively. However, fully understanding PPAR©’s role and how it controls insulin sensitivity is still elusive. Despite many side effects, which include heart failure, weight gain, bladder cancer, plasma volume expansion, and others, it is still considered an important target. Understanding its mechanism and exploring its downstream pathway is of great interest. Epigenetic modifications or post translational modification in PPAR©’s structure are thought to be promising routes in the treatment of various metabolic diseases. Various studies, including those on its phosphorylation at S273 or sumoylation at Lys107, have already demonstrated its importance in mediating insulin sensitizing actions [36]. Sirtuins-mediated changes in PPAR© also were beneficial in metabolic syndrome.

4.11. Glucocorticoid Receptor

The glucocorticoid receptor has a distinct effect on insulin action with both endogenous as well exogenous synthetic glucocorticoids [54]. In general, glucocorticoids increase the blood glucose levels when required by the brain, but chronic activation of these receptors is associated with metabolic dysfunction, including insulin resistance [36]. Glucocorticoids exhibit their effects either by directly acting on promoter or enhancer sites of the genes or by acting on other transcription factors and making a complex. Homodimerization and direct binding are associated with insulin resistance, affecting many gluconeogenic genes such as Pck1 and G6pc [36]. Chronic activation of these receptors inhibits protein synthesis and favors proteolysis. This ultimately releases amino acids and acts as substrate for glucose production. While the precise mechanism for how these receptors regulate insulin resistance is still unknown, many studies show that GR activation inhibits insulin stimulated glucose uptake by controlling many downstream targets.

4.12. Nuclear Factor (Erythroid-Derived 2)-Like 2 (NRF2)

NRF2 is an important molecular node which imparts cytoprotection in many diseases. Due to its different roles in various diseases, it is predicted to be an important target in drug discovery. With concern to type 2 patients with diabetes, a significant difference was found in the genotypic and allelic frequencies of four SNPs of the NFE2L2 gene. In addition, one study involved induction of NRF2 in obese diabetic db/db mice, displaying a decreased blood glucose level by suppressing hepatic glucose 6 phosphatase via cAMP-CREB signaling [55]. Leptin deficient mice showed decreased expression of NRF2 and displayed reduced amounts of white fat mass. This suggests that NRF2 is a key player in adipogenesis or in other metabolic disorders. It has been postulated that NRF2 levels can be increased with an acute dose of glucose, but chronic glucose conditions failed to activate it. Expression is also downregulated in prediabetes patients [55]. NRF2-mediated protection is well defined in the dysfunction of diabetic retinopathy as well. NRF2 knockdown enhances the palmitate-induced apoptosis in hepatocytes of obese patients.

4.13. Neprilysin

Neprilysin is a zinc-dependent metalloprotease that cleaves peptides (GLP-1 [7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36] amide, GLP-1 [9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36] amide from GLP-1 [28,29,30,31,32,33,34,35,36] amide and GLP-1 [32,33,34,35,36] amide) and inactivates several peptide hormones including glucagon, enkephalins, substance P, neurotensin, oxytocin, and bradykinin [56]. It was thought that GLP-1 [9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36] amide, which is the major circulation form in plasma, is the c-terminal metabolite which originates after the enzymatic cleavage of incretin hormone GLP-1 [7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36] amide by dipeptidyl peptidase-4. These new metabolites also were considered to have an insulinomimetic activity that contributes to the pleiotropic effects of GLP1, other than its canonical GLP-1R. It has been found that direct administration of these metabolites has antioxidant, anti-apoptotic, and proliferative effects on pancreatic β-cells by modulating energy homeostasis. It has been reported that these metabolites preserve mitochondrial function, inhibit membrane depolarization and caspase activation and hence, apoptosis of INS-1 β-cells. The effect of these metabolites on diabetes is further demonstrated by the administration of GLP-1 [28,29,30,31,32,33,34,35,36] amide, which increases β-cell mass and proliferation by activation of the cAMP/PKA signaling cascade and phosphorylation of the Wnt/β-catenin signaling pathway.

5. Drugs Targeting Molecular Pathways Implicated in Diabetes and CVD

In finding new therapeutics the multifactorial nature of T2DM should be considered, as well as prioritizing delivery of treatment at the earliest possible time. In addition, elevated risk of CVD is usually associated with T2DM. Consequently, current FDA guidelines necessitate that all new diabetic therapeutics should demonstrate protective or at least neutral cardiovascular impact profiles. In this way, targets of sub-atomic pathways conceivably involved in both diabetes and CVD are particularly attractive. Such methodologies incorporate the focusing of 11-hydroxysteroid dehydrogenase type 1 (11β-HSD1), GPR119, TGR5, sirtuin 1 (SIRT1), the sodium-glucose co-carrier 2 (SGLT2), and GPR40, for every one of which the reasoning is briefly depicted below (Table 3).
Taken together, these clinical findings give convincing evidence that various inhibitors, activators, agonists, and antagonists hold promise as new treatments for T2DM and related metabolic disorders.

6. Diabetes and the Gut Microbiota: Dietary Influence

Diet is a key factor influencing the composition of the gut microbiome, indicating the potential for therapeutic dietary strategies to manipulate microbial diversity, composition, and stability. Although changes in gut microbiota are temporary, diet can induce a shift in the gut microbiota. Numerous studies have pointed to a direct correlation between short-term dietary changes as well as long-term dietary changes with gut microbiome diversity in mouse models. In addition, it was found that gut microbiota participates in drug metabolism by producing specific enzymes, such as reductase and hydrolytic enzymes, thus affecting the efficacy, toxicity, and bioavailability of drugs. Various antibiotics also alter the diversity and composition of gut microbiota which leads to dysbiosis, and it was found to be linked with progression of various neurodegenerative as well as metabolic disorders [73,74,75].
Gut microbiota dysbiosis has been found as a major culprit in initiation as well as progression of various metabolic syndromes including diabetes mellitus, which unlocks various therapeutic interventions for the prevention as well as treatment of metabolic syndrome [76,77].
Isoflavone (genistein) reduced fasting glucose and insulin, as well as HOMA-IR, fibrinogen, and homocysteine levels [78]. However, the current study’s findings must be seen in the context of a healthy, well-balanced diet, which is likely to have boosted the positive benefits of genistein aglycone in our postmenopausal women [79]. Isoflavones have been shown to have anti-diabetic benefits due to their a-glucosidase inhibitory activity. It is thought that genistein prevents diabetes by preserving insulin-positive β-cells and altering the hepatic glucose metabolic enzyme profile. Genistein may prevent glucose-induced lipid peroxidation, modify cell islet pancreatic function, and increase basal metabolic rate and energy metabolism [80]. In vitro and in vivo studies have shown that genistein can serve as an estrogen agonist, resulting in the growth of estrogen-dependent human breast cancer tumors, and that long-term treatment with soy phytoestrogens has been linked to an increased risk of endometrial hyperplasia [81].

7. Crosstalk between Diabetes and Fatty Liver Disease-NASH and Prospective Therapeutic Drug Targets

Altered glucose and lipid metabolism are key factors associated with the development of NASH. Studies have shown the close relationship between metabolic disease and NASH [82,83]. People having NASH are more insulin resistant than healthy people, irrespective of their weight. Although most of the patients with NASH are insulin resistant, they do not show hyperglycemia because of decreased ability of the liver to clear insulin. NASH patients only develop diabetes when pancreatic beta cells are unable to increase insulin secretion to match the insulin resistance [84,85,86].
Some of the diabetic medications are used for the management and treatment of NASH. Glucagon-like-peptide-1 (GLP-1) receptors, which are used as second-line therapy for diabetes, have been used for amelioration of insulin resistance in NASH patients which ultimately improves the hepatic condition. The possible mechanism behind the improvement in insulin resistance is changes in body weight. However, researchers are still exploring the exact mechanism of these drugs [87,88]. Dipeptidyl peptidase (DPP)-4 inhibitors are another diabetic lowering class of agents that can be used for the treatment of NASH. Although these agents are not potent glucose-lowering agents due to their oral intake and low safety, these are used in combination with metformin in the management of diabetes. However, strong evidence is still missing for the clinical use of these agents for the management of NASH [89]. Pioglitazone, an anti-diabetic drug, has been shown to improve the condition in 60% of NASH patients. SGLT2 can be used for the treatment of NASH as it is helpful in glucose homeostasis. It has been shown to reverse liver steatosis and hepatocytes necrosis. These agents have been shown to decrease the plasma aminotransferases resulting in weight reduction which ultimately improves liver steatosis. One of these agents has been found to improve beta cell functions and insulin metabolism. Insulin has been under research for the treatment of liver steatosis and improvement in NASH [90].

8. Challenges and Opportunities

Despite the helpful impacts of current glucose-lowering drugs, morbidity and mortality remain significant in T2DM patients, highlighting the need for the development of pharmacotherapeutics with a special focus on the many metabolic irregularities and different pathways associated with morbidity. Probably the best test in T2DM treatment is to avoid the drawn-out side effects of any therapeutic intervention, including nonalcoholic greasy liver infection/nonalcoholic Steatohepatitis (NAFLD/NASH) and CVD. Forthcoming investigations in T2DM have demonstrated a relationship between the level of hyperglycemia and the danger of micro- and macro-vascular inconveniences, including deadly CVD events. In any case, the new ACCORD and ADVANCE findings in patients with longstanding T2DM have demonstrated that aggressive glucose control in such patients has no reasonable advantages or may even increase the risk of CVD events. In this manner, other independent risk factors may exist that together contribute to CVD risk in these patients. On the other hand, such discoveries may essentially mirror the restrictions of current diabetic treatments, due to off-target impacts that counter the likely advantages of glucose-lowering agents. The discovery of SGLT2 inhibitors that seemingly have only a few drawbacks needs to be further studied. In phase-III clinical trials, vulvovaginal candidiasis and mycotic infections were the most common side effects seen in females. Osmotic diuresis is possible as glucose excreted in the urine also increases water and solute output, which can further lead to ketoacidosis or acidosis by hyperkaliemia and renal insufficiency. On average, SGLT2 inhibitors are expensive and difficult to continue for long-term use (http://www.endocrinologynetwork.com/sglt2/sglt2-inhibitors-pros-cons-comparisons-and-considerations, 15 September 2021). Currently, the search for new drug molecules that can activate sirtuins is of peak interest, and here resveratrol is considered the best characterized compound. However, it does not activate SIRT1 directly. Many similar molecular structures have been made of SRT1720 and SRT2104, which are active. Still, they need some substrate, and surprisingly they activate some fluorophore tags. They activate SIRT1 directly if endogenous sirtuin is similar to the fluorophore substrate [91] (Figure 2).

9. Summary

The past thirty years have seen unrest in diabetes treatment optimization, especially in basic insulin treatment for glycemic control. This welcome activity is geared toward greater understanding of the hidden systems of disease pathology and the advancement of a great number of potential therapeutics. Lamentably, sedentary lifestyles and uneven dietary preferences have contributed to the current obesity pandemic. Along these lines, there is a steadily developing need to improve prediabetes and diabetes treatment approaches. While therapeutic options have unquestionably advanced, the International Diabetes Federation reports that many patients with diabetes remain unable or unwilling to follow current clinical care guidelines.
HbA1c-reducing agents are being evaluated in large clinical trials such as ACCORD, ADVANCE and VADT, in which improved control of HbA1c levels did result in reductions of serious microvascular diabetic complications, although rates of macrovascular infection and mortality remained essentially unchanged. It is consequently now understood that future therapeutic approaches should address clinical endpoints consistent with a multifactorial view of T2DM.
A wide variety of methodologies are being deployed to understand the multifactorial nature of T2DM. The ClinicalTrials.gov site records an excess of 1000 active type 2 diabetes trials around the world. These studies include research focused on cardiovascular involvement and liver diseases, among many others. By and large, the coming years hold great promise for the development of novel therapeutic interventions for T2DM and its related illnesses.

10. Future Perspectives

The medications currently available for T2DM have significant limitations and side effects. These notable shortcomings compromise their efficacy across many different clinical endpoints. Therefore, there is an ongoing need to search for new drug targets or combinations of existing drugs.
Future drug and therapeutic developments must follow a holistic approach with special focus on the following points: (1) they should target mechanisms to achieve efficient glycemic control with a safe weight loss profile; (2) they should mitigate the progression of diabetes-induced microvascular and cardiac complications.
Bariatric surgery has provided sensational improvements for many patients; however, significant relapse after six months has been reported. It is important to the scientific community to focus on the factors that may be involved in those early stages of improvement to advance T2DM research. To these ends, various groups have already started enrolling cohorts of obese diabetic and non-diabetic patients in a gastric banding trial. Blood and tissue samples will be collected from these patients (and a complementary control arm) at the time of the surgical procedure and then longitudinally during a five-year follow-up. The clinical and laboratory findings from this and other ongoing trials will no doubt contribute to advancing our therapeutic approaches to the management of diabetes and other metabolic disorders (Table 4).

Author Contributions

Conceptualization, S.P.S., methodology, P.S., N.K., S.B.; software, P.S., N.K., S.B.; validation, S.P.S., N.K., S.A. and A.K.; formal analysis, S.P.S.; investigation, A.K.; resources, P.S., N.K., S.B.; data curation, S.P.S.; writing—original draft preparation, S.P.S., S.B.; writing—review and editing, S.P.S., S.A., A.K.; visualization, A.K.; supervision, S.P.S.; project administration, S.P.S., S.A., A.K.; funding acquisition, S.A. All authors have read and agreed to the published version of the manuscript.

Funding

All authors have read and agree with the manuscript as written. SA is supported by grant support from NHLBI (RO1HL076801) and NIDCR (RO1DE014079).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

All authors have read and agree with the manuscript as written. The authors wish to thank Paola S. Salazar, M.A., and Gavin W. Hougham, for help with manuscript preparation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shrivastava, S.R.; Shrivastava, P.S.; Ramasamy, J. Role of self-care in management of diabetes mellitus. J. Diabetes Metab. Disord. 2013, 12, 14. [Google Scholar] [CrossRef] [Green Version]
  2. Sun, H.; Saeedi, P.; Karuranga, S.; Pinkepank, M.; Ogurtsova, K.; Duncan, B.B.; Stein, C.; Basit, A.; Chan, C.N.J.; Mbanya, C.J. IDF Diabetes Atlas: Global, regional, and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res. Clin. Pract. 2021, 109119. [Google Scholar] [CrossRef]
  3. Cassels, S. Overweight in the Pacific: Links between foreign dependence, global food trade, and obesity in the Federated States of Micronesia. Glob. Health 2006, 2, 10. [Google Scholar] [CrossRef] [Green Version]
  4. Chan, R.S.; Woo, J. Prevention of overweight and obesity: How effective is the current public health approach. Int. J. Environ. Res. Public Health 2010, 7, 765–783. [Google Scholar] [CrossRef] [Green Version]
  5. Ichiho, H.M.; Demei, Y.; Kuartei, S.; Aitaoto, N. An assessment of non-communicable diseases, diabetes, and related risk factors in the Republic of Palau: A systems perspective. Hawaii J. Med. Public Health 2013, 72 (Suppl. S1), 98–105. [Google Scholar]
  6. Kaveeshwar, S.A.; Cornwall, J. The current state of diabetes mellitus in India. Australas. Med. J. 2014, 7, 45–48. [Google Scholar] [CrossRef]
  7. Colberg, S.R.; Sigal, R.J.; Fernhall, B.; Regensteiner, J.G.; Blissmer, B.J.; Rubin, R.R.; Chasan-Taber, L.; Albright, A.L.; Braun, B. Exercise and type 2 diabetes: The American College of Sports Medicine and the American Diabetes Association: Joint position statement. Diabetes Care 2010, 33, e147–e167. [Google Scholar] [CrossRef] [Green Version]
  8. Alonso-Magdalena, P.; Ropero, A.B.; Soriano, S.; Quesada, I.; Nadal, A. Bisphenol-A: A new diabetogenic factor? Hormones 2010, 9, 118–126. [Google Scholar] [CrossRef]
  9. Lakhtakia, R. The History of Diabetes Mellitus. Sultan Qaboos Univ. Med. J. 2013, 13, 368–370. [Google Scholar] [CrossRef]
  10. Nejabat, M.; Maleki, B.; Nimrouzi, M.; Mahbodi, A.; Salehi, A. Avicenna and Cataracts: A New Analysis of Contributions to Diagnosis and Treatment from the Canon. Iran. Red Crescent Med. J. 2012, 14, 265–270. [Google Scholar]
  11. Tan, S.Y.; Merchant, J. Frederick Banting (1891–1941): Discoverer of insulin. Singap. Med. J. 2017, 58, 2–3. [Google Scholar] [CrossRef]
  12. WH Organization. Classification of Diabetes Mellitus. 2019. Available online: https://www.who.int/publications/i/item/classification-of-diabetes-mellitus (accessed on 15 October 2021).
  13. Quinn, L.M.; Wong, F.S.; Narendran, P. Environmental Determinants of Type 1 Diabetes: From Association to Proving Causality. Front. Immunol. 2021, 12, 737964. [Google Scholar] [CrossRef]
  14. Rui, L. Energy metabolism in the liver. Compr. Physiol. 2014, 4, 177–197. [Google Scholar] [CrossRef] [Green Version]
  15. Williams, A.S.; Kang, L.; Wasserman, D.H. The extracellular matrix and insulin resistance. Trends Endocrinol. Metab. 2015, 26, 357–366. [Google Scholar] [CrossRef] [Green Version]
  16. Herath, H.; Herath, R.; Wickremasinghe, R. Gestational diabetes mellitus and risk of type 2 diabetes 10 years after the index pregnancy in Sri Lankan women-A community based retrospective cohort study. PLoS ONE 2017, 12, e0179647. [Google Scholar] [CrossRef] [Green Version]
  17. Pilacinski, S.; Zozulinska-Ziolkiewicz, D.A. Influence of lifestyle on the course of type 1 diabetes mellitus. Arch. Med. Sci. 2014, 10, 124–134. [Google Scholar] [CrossRef] [Green Version]
  18. Moller David, E. Metabolic Disease Drug Discovery—“Hitting the Target” Is Easier Said Than Done. Cell Metab. 2012, 15, 19–24. [Google Scholar] [CrossRef] [Green Version]
  19. Ngoc Doan Trang, N.; Ly Thi, L. Targeted proteins for diabetes drug design. Adv. Nat. Sci. Nanosci. Nanotechnol. 2012, 3, 013001. [Google Scholar] [CrossRef]
  20. Kunhiraman, B.P.; Jawa, A.; Fonseca, V.A. Potential cardiovascular benefits of insulin sensitizers. Endocrinol. Metab. Clin. N. Am. 2005, 34, 117–135. [Google Scholar] [CrossRef]
  21. McCarty, M.F.; DiNicolantonio, J.J. Acarbose, lente carbohydrate, and prebiotics promote metabolic health and longevity by stimulating intestinal production of GLP-1. Open Heart 2015, 2, e000205. [Google Scholar] [CrossRef]
  22. Nauck, M. Incretin therapies: Highlighting common features and differences in the modes of action of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Diabetes Obes. Metab. 2016, 18, 203–216. [Google Scholar] [CrossRef] [Green Version]
  23. Mikhail, N. Place of sodium-glucose co-transporter type 2 inhibitors for treatment of type 2 diabetes. World J. Diabetes 2014, 5, 854–859. [Google Scholar] [CrossRef]
  24. Cooper, M.S.; Stewart, P.M. 11Beta-hydroxysteroid dehydrogenase type 1 and its role in the hypothalamus-pituitary-adrenal axis, metabolic syndrome, and inflammation. J. Clin. Endocrinol. Metab. 2009, 94, 4645–4654. [Google Scholar] [CrossRef] [Green Version]
  25. Schleicher, E.D.; Weigert, C. Role of the hexosamine biosynthetic pathway in diabetic nephropathy. Kidney Int. 2000, 58, S13–S18. [Google Scholar] [CrossRef] [Green Version]
  26. Stull, A.J.; Wang, Z.Q.; Zhang, X.H.; Yu, Y.; Johnson, W.D.; Cefalu, W.T. Skeletal muscle protein tyrosine phosphatase 1B regulates insulin sensitivity in African Americans. Diabetes 2012, 61, 1415–1422. [Google Scholar] [CrossRef] [Green Version]
  27. Fernandez-Ruiz, R.; Vieira, E.; Garcia-Roves, P.M.; Gomis, R. Protein tyrosine phosphatase-1B modulates pancreatic beta-cell mass. PLoS ONE 2014, 9, e90344. [Google Scholar] [CrossRef] [Green Version]
  28. Qian, S.; Zhang, M.; He, Y.; Wang, W.; Liu, S. Recent advances in the development of protein tyrosine phosphatase 1B inhibitors for Type 2 diabetes. Future Med. Chem. 2016, 8, 1239–1258. [Google Scholar] [CrossRef]
  29. Rusu, V.; Hoch, E.; Mercader, J.M.; Tenen, D.E.; Gymrek, M.; Hartigan, C.R.; DeRan, M.; von Grotthuss, M.; Fontanillas, P.; Spooner, A.; et al. Type 2 Diabetes Variants Disrupt Function of SLC16A11 through Two Distinct Mechanisms. Cell 2017, 170, 199–212. [Google Scholar] [CrossRef] [Green Version]
  30. Rhee, E.P.; Cheng, S.; Larson, M.G.; Walford, G.A.; Lewis, G.D.; McCabe, E.; Yang, E.; Farrell, L.; Fox, C.S.; O’Donnell, C.J.; et al. Lipid profiling identifies a triacylglycerol signature of insulin resistance and improves diabetes prediction in humans. J. Clin. Investig. 2011, 121, 1402–1411. [Google Scholar] [CrossRef] [Green Version]
  31. Samuel, V.T.; Shulman, G.I. Mechanisms for insulin resistance: Common threads and missing links. Cell 2012, 148, 852–871. [Google Scholar] [CrossRef] [Green Version]
  32. Lin, Z.; Natesan, V.; Shi, H.; Hamik, A.; Kawanami, D.; Hao, C.; Mahabaleshwar, G.H.; Wang, W.; Jin, Z.G.; Atkins, G.B.; et al. A novel role of CCN3 in regulating endothelial inflammation. J. Cell Commun. Signal. 2010, 4, 141–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Pakradouni, J.; Le Goff, W.; Calmel, C.; Antoine, B.; Villard, E.; Frisdal, E.; Abifadel, M.; Tordjman, J.; Poitou, C.; Bonnefont-Rousselot, D.; et al. Plasma NOV/CCN3 levels are closely associated with obesity in patients with metabolic disorders. PLoS ONE 2013, 8, e66788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Martinerie, C.; Garcia, M.; Do, T.T.; Antoine, B.; Moldes, M.; Dorothee, G.; Kazazian, C.; Auclair, M.; Buyse, M.; Ledent, T.; et al. NOV/CCN3: A New Adipocytokine Involved in Obesity-Associated Insulin Resistance. Diabetes 2016, 65, 2502–2515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Li, J.Y.; Wang, Y.D.; Qi, X.Y.; Ran, L.; Hong, T.; Yang, J.; Yan, B.; Liao, Z.Z.; Liu, J.H.; Xiao, X.H. Serum CCN3 levels are increased in type 2 diabetes mellitus and associated with obesity, insulin resistance and inflammation. Clin. Chim. Acta 2019, 494, 52–57. [Google Scholar] [CrossRef]
  36. Kang, S.; Tsai, L.T.; Rosen, E.D. Nuclear Mechanisms of Insulin Resistance. Trends Cell Biol. 2016, 26, 341–351. [Google Scholar] [CrossRef] [Green Version]
  37. Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.Y.; Lannoy, V.; Decobecq, M.E.; Brezillon, S.; Dupriez, V.; Vassart, G.; van Damme, J.; et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef] [Green Version]
  38. Nilsson, N.E.; Kotarsky, K.; Owman, C.; Olde, B. Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochem. Biophys. Res. Commun. 2003, 303, 1047–1052. [Google Scholar] [CrossRef]
  39. Li, M.; van Esch, B.; Wagenaar, G.T.M.; Garssen, J.; Folkerts, G.; Henricks, P.A.J. Pro- and anti-inflammatory effects of short chain fatty acids on immune and endothelial cells. Eur. J. Pharmacol. 2018, 831, 52–59. [Google Scholar] [CrossRef]
  40. Kebede, M.A.; Alquier, T.; Latour, M.G.; Poitout, V. Lipid receptors and islet function: Therapeutic implications? Diabetes Obes. Metab. 2009, 11 (Suppl. S4), 10–20. [Google Scholar] [CrossRef] [Green Version]
  41. Bahar Halpern, K.; Veprik, A.; Rubins, N.; Naaman, O.; Walker, M.D. GPR41 gene expression is mediated by internal ribosome entry site (IRES)-dependent translation of bicistronic mRNA encoding GPR40 and GPR41 proteins. J. Biol. Chem. 2012, 287, 20154–20163. [Google Scholar] [CrossRef] [Green Version]
  42. den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Tang, C.; Ahmed, K.; Gille, A.; Lu, S.; Gröne, H.J.; Tunaru, S.; Offermanns, S. Loss of FFA2 and FFA3 increases insulin secretion and improves glucose tolerance in type 2 diabetes. Nat. Med. 2015, 21, 173–177. [Google Scholar] [CrossRef] [PubMed]
  44. Karamanou, M.; Protogerou, A.; Tsoucalas, G.; Androutsos, G.; Poulakou-Rebelakou, E. Milestones in the history of diabetes mellitus: The main contributors. World J. Diabetes 2016, 7, 1–7. [Google Scholar] [CrossRef] [PubMed]
  45. Capdevila, J.H.; Falck, J.R.; Harris, R.C. Cytochrome P450 and arachidonic acid bioactivation. Molecular and functional properties of the arachidonate monooxygenase. J. Lipid Res. 2000, 41, 163–181. [Google Scholar] [CrossRef]
  46. Waldman, M.; Bellner, L.; Vanella, L.; Schragenheim, J.; Sodhi, K.; Singh, S.P.; Lin, D.; Lakhkar, A.; Li, J.; Hochhauser, E.; et al. Epoxyeicosatrienoic Acids Regulate Adipocyte Differentiation of Mouse 3T3 Cells, Via PGC-1alpha Activation, Which Is Required for HO-1 Expression and Increased Mitochondrial Function. Stem Cells Dev. 2016, 25, 1084–1094. [Google Scholar] [CrossRef] [Green Version]
  47. Zeldin, D.C. Epoxygenase pathways of arachidonic acid metabolism. J. Biol. Chem. 2001, 276, 36059–36062. [Google Scholar] [CrossRef] [Green Version]
  48. Node, K.; Huo, Y.; Ruan, X.; Yang, B.; Spiecker, M.; Ley, K.; Zeldin, D.C.; Liao, J.K. Anti-inflammatory properties of cytochrome P450 epoxygenase-derived eicosanoids. Science 1999, 285, 1276–1279. [Google Scholar] [CrossRef] [Green Version]
  49. Spiecker, M.; Liao, J.K. Vascular protective effects of cytochrome p450 epoxygenase-derived eicosanoids. Arch. Biochem. Biophys. 2005, 433, 413–420. [Google Scholar] [CrossRef]
  50. Zha, W.; Edin, M.L.; Vendrov, K.C.; Schuck, R.N.; Lih, F.B.; Jat, J.L.; Bradbury, J.A.; DeGraff, L.M.; Hua, K.; Tomer, K.B.; et al. Functional characterization of cytochrome P450-derived epoxyeicosatrienoic acids in adipogenesis and obesity. J. Lipid Res. 2014, 55, 2124–2136. [Google Scholar] [CrossRef] [Green Version]
  51. Wu, Z.; Puigserver, P.; Andersson, U.; Zhang, C.; Adelmant, G.; Mootha, V.; Troy, A.; Cinti, S.; Lowell, B.; Scarpulla, R.C.; et al. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell 1999, 98, 115–124. [Google Scholar] [CrossRef] [Green Version]
  52. Wenz, T.; Rossi, S.G.; Rotundo, R.L.; Spiegelman, B.M.; Moraes, C.T. Increased muscle PGC-1alpha expression protects from sarcopenia and metabolic disease during aging. Proc. Natl. Acad. Sci. USA 2009, 106, 20405–20410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kleiner, S.; Mepani, R.J.; Laznik, D.; Ye, L.; Jurczak, M.J.; Jornayvaz, F.R.; Estall, J.L.; Chatterjee Bhowmick, D.; Shulman, G.I.; Spiegelman, B.M.; et al. Development of insulin resistance in mice lacking PGC-1alpha in adipose tissues. Proc. Natl. Acad. Sci. USA 2012, 109, 9635–9640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Moraitis, A.G.; Block, T.; Nguyen, D.; Belanoff, J.K. The role of glucocorticoid receptors in metabolic syndrome and psychiatric illness. J. Steroid Biochem. Mol. Biol. 2017, 165 Pt A, 114–120. [Google Scholar] [CrossRef] [Green Version]
  55. Cuadrado, A.; Manda, G.; Hassan, A.; Alcaraz, M.J.; Barbas, C.; Daiber, A.; Ghezzi, P.; León, R.; López, M.G.; Oliva, B.; et al. Transcription Factor NRF2 as a Therapeutic Target for Chronic Diseases: A Systems Medicine Approach. Pharm. Rev. 2018, 70, 348–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Guglielmi, V.; Sbraccia, P. GLP-1 receptor independent pathways: Emerging beneficial effects of GLP-1 breakdown products. Eat. Weight Disord. 2017, 22, 231–240. [Google Scholar] [CrossRef]
  57. Hamilton, B.S.; Himmelsbach, F.; Nar, H.; Schuler-Metz, A.; Krosky, P.; Guo, J.; Guo, R.; Meng, S.; Zhao, Y.; Lala, D.S.; et al. Pharmacological characterization of the selective 11β-hydroxysteroid dehydrogenase 1 inhibitor, BI 135585, a clinical candidate for the treatment of type 2 diabetes. Eur. J. Pharmacol. 2015, 746, 50–55. [Google Scholar] [CrossRef]
  58. Martocchia, A.; Stefanelli, M.; Falaschi, G.M.; Toussan, L.; Ferri, C.; Falaschi, P. Recent advances in the role of cortisol and metabolic syndrome in age-related degenerative diseases. Aging Clin. Exp. Res. 2016, 28, 17–23. [Google Scholar] [CrossRef] [PubMed]
  59. Yu, D.D.; Sousa, K.M.; Mattern, D.L.; Wagner, J.; Fu, X.; Vaidehi, N.; Forman, B.M.; Huang, W. Stereoselective synthesis, biological evaluation, and modeling of novel bile acid-derived G-protein coupled Bile acid receptor 1 (GP-BAR1, TGR5) agonists. Bio Organ. Med. Chem. 2015, 23, 1613–1628. [Google Scholar] [CrossRef]
  60. Ido, Y. Diabetic complications within the context of ageing: NADH/NAD+ redox, insulin C-peptide, SIRT1-LKB1-AMPK positive feedback and FOXO3. J. Diabetes Investig. 2016, 7, 448–458. [Google Scholar] [CrossRef]
  61. Kanwal, A.; Banerjee, S.K. SGLT inhibitors: A novel target for diabetes. Pharm. Pat. Anal. 2013, 2, 77–91. [Google Scholar] [CrossRef]
  62. Bailey, C.J.; Tahrani, A.A.; Barnett, A.H. Future glucose-lowering drugs for type 2 diabetes. Lancet Diabetes Endocrinol. 2016, 4, 350–359. [Google Scholar] [CrossRef] [Green Version]
  63. Malek, R.; Davis, S.N. Tyrosine kinase inhibitors under investigation for the treatment of type II diabetes. Expert Opin. Investig. Drugs 2016, 25, 287–296. [Google Scholar] [CrossRef]
  64. Tang, W.J. Targeting Insulin-Degrading Enzyme to Treat Type 2 Diabetes Mellitus. Trends Endocrinol. Metab. 2016, 27, 24–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Rosqvist, F.; Iggman, D.; Kullberg, J.; Cedernaes, J.; Johansson, H.E.; Larsson, A.; Johansson, L.; Ahlström, H.; Arner, P.; Dahlman, I.; et al. Overfeeding polyunsaturated and saturated fat causes distinct effects on liver and visceral fat accumulation in humans. Diabetes 2014, 63, 2356–2368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Tao, R.; Xiong, X.; Liangpunsakul, S.; Dong, X.C. Sestrin 3 Protein Enhances Hepatic Insulin Sensitivity by Direct Activation of the mTORC2-Akt Signaling. Diabetes 2014, 64, 1211–1223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Achari, A.E.; Jain, S.K. Adiponectin, a Therapeutic Target for Obesity, Diabetes, and Endothelial Dysfunction. Int. J. Mol. Sci. 2017, 18, 1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Okada-Iwabu, M.; Yamauchi, T.; Iwabu, M.; Honma, T.; Hamagami, K.I.; Matsuda, K.; Yamaguchi, M.; Tanabe, H.; Kimura-Someya, T.; Shirouzu, M.; et al. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature 2013, 503, 493–499. [Google Scholar] [CrossRef]
  69. Li, Y.; Zheng, L.; Wang, D.; Zhang, X.; Li, J.; Ali, S.; Lu, J.; Zong, H.; Xu, X. Staurosporine as an agonist for induction of GLUT4 translocation, identified by a pH-sensitive fluorescent IRAP-mOrange2 probe. Biochem. Biophys. Res. Commun. 2016, 480, 534–538. [Google Scholar] [CrossRef]
  70. Zhang, Y.; Zhang, H.; Yao, X.G.; Shen, H.; Chen, J.; Li, C.; Chen, L.; Zheng, M.; Ye, J.; Hu, L.; et al. (+)-Rutamarin as a dual inducer of both GLUT4 translocation and expression efficiently ameliorates glucose homeostasis in insulin-resistant mice. PLoS ONE 2012, 7, e31811. [Google Scholar] [CrossRef] [Green Version]
  71. Liang, H.; Ward, W.F. PGC-1alpha: A key regulator of energy metabolism. Adv. Physiol. Educ. 2006, 30, 145–151. [Google Scholar] [CrossRef]
  72. Woundman, Y.T. Obesity, Insulin Resistance, and Type 2 Diabetes: Associations and Therapeutic Implications. Diabetes Metab. Syndr. Obes. 2020, 13, 3611–3616. [Google Scholar] [CrossRef] [PubMed]
  73. David, L.; Materna, A.; Friedman, J.; Campos-Baptista, M.; Blackburn, M.; Perrotta, A.; Erdman, S.E.; Alm, E.J. Host lifestyle affects human microbiota on daily timescales. Genome Biol. 2014, 15, R89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Díez-Sainz, E.; Milagro, F.; Riezu-Boj, J.; Lorente-Cebrián, S. Effects of gut microbiota–derived extracellular vesicles on obesity and diabetes and their potential modulation through diet. J. Physiol. Biochem. 2021. [Google Scholar] [CrossRef] [PubMed]
  75. Leeming, E.; Johnson, A.; Spector, T.; Le Roy, C. Effect of Diet on the Gut Microbiota: Rethinking Intervention Duration. Nutrients 2019, 11, 2862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wu, G.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.; Keilbaugh, S.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Xie, Y.; Hu, F.; Xiang, D.; Lu, H.; Li, W.; Zhao, A.; Huang, L.; Wang, R. The metabolic effect of gut microbiota on drugs. Drug Metab. Rev. 2020, 52, 139–156. [Google Scholar] [CrossRef]
  78. Maliehe, A.; Ghahremani, S.; Kharghani, S.; Masumeh, G.; Maliehe, A.; Ghahremani, S.; Kharghani, S.; Ghazanfarpour, M.; Shariati, K.; Kazemi, M.; et al. Effect of Isoflavones and Genistein on Glucose Metabolism in Peri- and Post-Menopausal Women: An Overview of Meta-Analysis. J. Menopausal Med. 2019, 25, 69–73. [Google Scholar] [CrossRef]
  79. Squadrito, F.; Marini, H.; Bitto, A.; Altavilla, D.; Polito, F.; Adamo, E.B.; D’Anna, R.; Arcoraci, V.; Burnett, B.P.; Minutoli, L.; et al. Genistein in the Metabolic Syndrome: Results of a Randomized Clinical Trial. J. Clin. Endocrinol. Metab. 2013, 98, 3366–3374. [Google Scholar] [CrossRef] [Green Version]
  80. Ma, L.; Liu, G.; Ding, M.; Zong, G.; Hu, F.B.; Willett, W.C.; Rimm, E.B.; Manson, J.E.; Sun, Q. Isoflavone Intake and the Risk of Coronary Heart Disease in US Men and Women. Circulation 2020, 141, 1127–1137. [Google Scholar] [CrossRef]
  81. Omoruyi, F.; Stennett, D.; Foster, S.; Dilworth, L. New Frontiers for the Use of IP6 and Inositol Combination in Treating Diabetes Mellitus: A Review. Molecules 2020, 25, 1720. [Google Scholar] [CrossRef] [Green Version]
  82. Yki-Jarvinen, H. Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome. Lancet Diabetes Endocrinol. 2014, 2, 901–910. [Google Scholar] [CrossRef]
  83. Vernon, G.; Baranova, A.; Younossi, Z.M. Systematic review: The epidemiology and natural history of non-alcoholic fatty liver disease and nonalcoholic steatohepatitis in adults. Aliment. Pharm. Ther. 2011, 34, 274–285. [Google Scholar] [CrossRef] [PubMed]
  84. Bugianesi, E.; Gastaldelli, A.; Vanni, E.; Gambino, R.; Cassader, M.; Baldi, S.; Ponti, V.; Pagano, G.; Ferrannini, E.; Rizzetto, M. Insulin resistance in non-diabetic patients with non-alcoholic fatty liver disease: Sites and mechanisms. Diabetologia 2005, 48, 634–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Gastaldelli, A.; Cusi, K.; Pettiti, M.; Hardies, J.; Miyazaki, Y.; Berria, R.; Buzzigoli, E.; Sironi, A.M.; Cersosimo, E.; Ferrannini, E.; et al. Relationship between hepatic/visceral fat and hepatic insulin resistance in nondiabetic and type 2 diabetic subjects. Gastroenterology 2007, 133, 496–506. [Google Scholar] [CrossRef]
  86. Bril, F.; Lomonaco, R.; Orsak, B.; Ortiz-Lopez, C.; Webb, A.; Tio, F.; Hecht, J.; Cusi, K. Relationship between disease severity, hyperinsulinemia, and impaired insulin clearance in patients with nonalcoholic steatohepatitis. Hepatology 2014, 59, 2178–2187. [Google Scholar] [CrossRef]
  87. Cersosimo, E.; Gastaldelli, A.; Cervera, A.; Wajcberg, E.; Sriwijilkamol, A.; Fernandez, M.; Zuo, P. Effect of exenatide on splanchnic and peripheral glucose metabolism in type 2 diabetic subjects. J. Clin. Endocrinol. Metab. 2011, 96, 1763–1770. [Google Scholar] [CrossRef]
  88. Armstrong, M.J.; Hull, D.; Guo, K.; Barton, D.; Hazlehurst, J.M.; Gathercole, L.L.; Nasiri, M.; Yu, J.; Gough, S.C.; Newsome, P.N.; et al. Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis. J. Hepatol. 2016, 64, 399–408. [Google Scholar] [CrossRef] [Green Version]
  89. Scheen, A. The safety of gliptins: Updated data in 2018. Expert Opin. Drug Saf. 2018, 17, 387–405. [Google Scholar] [CrossRef]
  90. Cusi, K.; Sanyal, A.J.; Zhang, S.; Hartman, M.L.; Bue-Valleskey, J.M.; Hoogwerf, B.J.; Haupt, A. Nonalcoholic fatty liver disease (NAFLD) prevalence and its metabolic associations in patients with type 1 diabetes and type 2 diabetes. Diabetes Obes. Metab. 2017, 19, 1630–1634. [Google Scholar] [CrossRef]
  91. Andreux, P.A.; Houtkooper, R.H.; Auwerx, J. Pharmacological approaches to restore mitochondrial function. Review. Nat. Rev. Drug Discov. 2013, 12, 465–483. [Google Scholar] [CrossRef] [Green Version]
Figure 1. (a) Sodium-glucose co-transporter type 2 (SGLT2) inhibitor pathway. (b) Sodium-glucose co-transporter type 2 (SGLT2) inhibitor pathway.
Figure 1. (a) Sodium-glucose co-transporter type 2 (SGLT2) inhibitor pathway. (b) Sodium-glucose co-transporter type 2 (SGLT2) inhibitor pathway.
Biomedicines 10 00331 g001
Figure 2. Timeline of t2DM Drug Development.
Figure 2. Timeline of t2DM Drug Development.
Biomedicines 10 00331 g002
Table 1. World diabetes burden by year 2045.
Table 1. World diabetes burden by year 2045.
2019 RankCountry/Territory2019
(Millions)
Country/Territory2045 (Millions)
1China116.4India134.3
2India77China119.8
3U. S. A.31U. S. A.35.6
4Pakistan19.4Mexico21.8
5Brazil16.8Brazil20.3
6Mexico12.8Egypt16.7
7Indonesia10.7Indonesia16.7
8Germany9.5Pakistan16.1
9Egypt8.9Bangladesh13.7
10Bangladesh8.4Turkey11.2
Table 2. New anti-diabetic drugs in the drug discovery pipeline.
Table 2. New anti-diabetic drugs in the drug discovery pipeline.
NameSponsor/DeveloperMechanism of ActionIndication
AfrezzaMannKindUltra-rapid-acting mealtime insulin therapyAdults with type 1 or type 2 diabetes
Albiglutide GlaxoSmithKlineGlucagon-like peptide (GLP) 1 agonistOnce weekly for adults with type 2 diabetes
Aleglitazar RocheDual peroxisome proliferator-activated receptor (PPAR) α/γ activationCardiovascular risk reduction in type 2 diabetes
Alogliptin, Alogliptin and pioglitazone, Alogliptin and metformin Takeda Pharmaceuticals and Furiex PharmaceuticalsDPP-4 inhibitorOral treatment of type 2 diabetes, individually and in two fixed-dose combinations
AtrasentanAbbVieSelective endothelin-A receptor antagonistOral once-daily treatment for diabetic nephropathy
Dulaglutide (LY2189265)Eli LillyGLP-1 analogOnce weekly for type 2 diabetes
Empagliflozin (BI10773)Boehringer Ingelheim and Eli LillySodium dependent glucose transporter 2 (SGLT2) inhibitorOral treatment for adults with type 2 diabetes
Ertugliflozin (MK-8835; PF-04971729)Merck & Co., licensed from PfizerSGLT2 inhibitorType 2 diabetes
Fasiglifam (TAK-875)TakedaG-protein-coupled receptor (GPCR) 40 agonistType 2 diabetes
FIAsp (NN1218)Novo NordiskFaster-acting formulation of insulin aspartType 1 and 2 diabetes
Forxiga™ (dapagliflozin)Bristol Myers Squibb and AstraZenecaSGLT2 inhibitorOnce-daily tablets for adults with type 2 diabetes
IDegLira (NN9068)Novo NordiskCombination drug therapyType 2 diabetes
Invokana (canagliflozin)Johnson & JohnsonSGLT2 inhibitorOnce-daily tablets for adults with type 2 diabetes
Ipragliflozin L-proline (ASP1941)Astellas, MSD, and Kotobuki PharmaceuticalSGLT2 inhibitorType 2 diabetes
Luseogliflozin hydrate (TS-071)Taisho PharmaceuticalSGLT2 inhibitorOnce-daily for type 2 diabetes
LixiLan (lixisenatide+ insulin glargine)Sanofi; lixisenatideCombination drug therapy Type 2 diabetes
Lyxumia® (lixisenatide)Sanofi; licensed from Zealand PharmaGLP-1 agonistOnce-daily for type 2 diabetes
LY2605541 (basal insulin peglispro)Eli LillyBasal insulin analogType 1 and 2 diabetes
LY2963016 (new insulin glargine product)Eli Lilly and Boehringer IngelheimBasal insulinType 1 and 2 diabetes
Omarigliptin (MK-3102)Merck & Co.DPP-4 inhibitorOnce-weekly for adults with type 2 diabetes
Ryzodeg® (insulin degludec + insulin aspart)Novo NordiskSoluble fixed combination of basal insulin with bolus insulin aspart Once-daily for type 1 and 2 diabetes
Semaglutide (NN9535)Novo NordiskGLP-1 analogOnce-weekly for type 2 diabetes
SYR-472 (trelagliptin succinate)Takeda Pharmaceuticals and Furiex PharmaceuticalsDPP-4 inhibitorOnce-weekly oral treatment for type 2 diabetes
Tresiba® (Insulin degludec)Novo NordiskOnce-daily basal insulinType 1 and 2 diabetes
U300SanofiInsulin glargineType 1 and 2 diabetes
Table 3. List of new anti-diabetic drugs targeting molecular pathways implicated in diabetes and CVD.
Table 3. List of new anti-diabetic drugs targeting molecular pathways implicated in diabetes and CVD.
TargetsMechanism of ActionLeadsRef.
11β-HSD1
(hydroxysteroid dehydrogenase)
Blocking cortisol1. INCB13739, MK-0916,
2. BI 135585
[57,58]
G protein-coupled receptor (GPR119) Increases cAMP signalingAPD5979, MBX-2982
TGR5
(bile-acid activated GPCR)
cAMP accumulation and enhances GLP-1 secretion (intestine), anti-inflammatory effect (liver) INT-777[59]
SIRT1Improves insulin sensitivity, increases glucose homeostasis, increases mitochondrial capacitySRT2104, resveratrol[60]
SGLT-2Increases kidney-dependent glucose homeostasisDapagliflozin, canagliflozin, sergliflozin, remogliflozin, ipragliflozin, and empagliflozin, etc.[61]
GPR40Increases incretin secretion, improves glucose toleranceModulators [62]
PPAR-γImproves serum lipid profile, glucose homeostasis, insulin sensitivity, reduces inflammation and weight gainPiolitazone, aleglitazar, glitazones, GFT505
Tyrosine KinaseReduces beta cell apoptosis, enhances insulin secretion, increases beta cell survival, reduces insulin resistance, Imatinib, Sunitinib, Dasatinib, Sorafenib, Erlotinib[63]
PPLR Improves JAK2/STAT5 pathway for glucose uptakeBromocriptine
Insulin degrading enzyme (IDE)Thiol zinc-metalloendopeptidase that cleaves small proteins of diverse sequenceBDM44768, 6bK, NTE-1[64]
FATP5Enhances the uptake of long-chain and very long-chain fatty acids into the cellsChenodiol and Ursodiol [65]
SestrinEnhances hepatic insulin sensitivity3. Enhances hepatic insulin sensitivity[66]
StatinFor diabetic dyslipidemia
AdiponectinDecreased adiponectin levels are thought to play
a central role in the development of type 2 diabetes and obesity
AdipoRon is a novel orally-active small molecule that serves as a potent selective agonist of the AdipoR1 and AdipoR2 adiponectin receptors[67,68]
Glut4Triggering the canonical PI3K–AKT pathway is essential and ample to activate exocytosis of GLUT4 storage vesicles to the plasma membraneStaurosporine is used to promotes GSVs translocation and glucose uptake through the AMPK pathway
Rutamarin as a dual inducer of both GLUT4 translocation and expression efficiently ameliorates glucose homeostasis in insulin-resistant mice
[69,70]
PGC-1αReduction in PGC-1α triggers insulin resistance and ultimately causes diabetesStill waiting to synthesize agonist[71,72]
Table 4. Diabetes Mellitus.
Table 4. Diabetes Mellitus.
Trend Box
Diabetes mellitus is a complex metabolic disruption characterized by chronic hyperglycemia due to deficiencies in insulin production, its systemic release and action, and resistance.
Incident diabetes is also due to changes in performance of multiple key enzymes. Therefore, identifying and understanding key targets at structure-function-dynamics levels and their potential leads and/or drugs, shortcomings, and associated challenges all together can provide a roadmap for the discovery of new selective modulators.
We listed promising therapeutic targets for treatment of T2DM, most of them involving improvements in glucose tolerance.
Novel drugs from various targets have numerous locales of activity and each focus on a few components of the metabolic disorder. These may help pave the way for future type 2 diabetes treatments.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kanwal, A.; Kanwar, N.; Bharati, S.; Srivastava, P.; Singh, S.P.; Amar, S. Exploring New Drug Targets for Type 2 Diabetes: Success, Challenges and Opportunities. Biomedicines 2022, 10, 331. https://doi.org/10.3390/biomedicines10020331

AMA Style

Kanwal A, Kanwar N, Bharati S, Srivastava P, Singh SP, Amar S. Exploring New Drug Targets for Type 2 Diabetes: Success, Challenges and Opportunities. Biomedicines. 2022; 10(2):331. https://doi.org/10.3390/biomedicines10020331

Chicago/Turabian Style

Kanwal, Abhinav, Navjot Kanwar, Sanjay Bharati, Prateek Srivastava, Shailendra P. Singh, and Salomon Amar. 2022. "Exploring New Drug Targets for Type 2 Diabetes: Success, Challenges and Opportunities" Biomedicines 10, no. 2: 331. https://doi.org/10.3390/biomedicines10020331

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop