IMR Press / FBL / Volume 28 / Issue 7 / DOI: 10.31083/j.fbl2807134
Open Access Review
SGLT-2 Inhibitors for Non-Alcoholic Fatty Liver Disease: A Review
Rong Xu1,2,†Difei Lian1,2,†Yan Xie1,2Zhilei Chen1,2Yan Wang1,2Lin Mu1,2Yuan Wang1,2Baoyu Zhang1,2,*
Show Less
1 Center for Endocrine Metabolism and Immune Diseases, Lu He Hospital, Capital Medical University, 100000 Beijing, China
2 Beijing Key Laboratory of Diabetes Research and Care, 100000 Beijing, China
*Correspondence: lhyynfm@126.com (Baoyu Zhang)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(7), 134; https://doi.org/10.31083/j.fbl2807134
Submitted: 28 December 2022 | Revised: 16 February 2023 | Accepted: 13 March 2023 | Published: 6 July 2023
(This article belongs to the Special Issue An Update on Sodium Glucose Co-Transporters)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Non-alcoholic fatty liver disease (NAFLD) is a group of metabolic liver illnesses that lead to accumulation of liver fat mainly due to excessive nutrition. It is closely related to insulin resistance, obesity, type 2 diabetes, and cardiovascular disease, and has become one of the main causes of chronic liver disease worldwide. At present, there is no specific drug for the treatment of NAFLD; lifestyle interventions including dietary control and exercise are recommended as routine treatments. As a drug for the treatment of type 2 diabetes, sodium-glucose co-transporter type 2 (SGLT-2) inhibitors may also play a beneficial role in the treatment of NAFLD. This article reviews the mechanism of SGLT-2 inhibitors in the treatment of NAFLD.

Keywords
SGLT-2 inhibitors
non-alcoholic fatty liver disease
non-alcoholic steatohepatitis
type 2 diabetes
1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is a group of liver diseases associated with metabolic dysfunction, such as diffuse non-alcoholic liver steatosis, non-alcoholic steatohepatitis (NASH), and other features of liver damage, such as liver cirrhosis and hepatocellular carcinoma (HCC) [1]. NAFLD is defined as the presence of steatosis in more than 5% of hepatocytes and the absence of causes like excessive alcohol consumption, drugs, or other chronic liver diseases. NAFLD has become one of the main reasons for chronic liver disease around the world. The prevalence of NAFLD is reported to be about 25% worldwide, ranging from 13.5% in Africa to 31.8% in the Middle East [2, 3].

NAFLD has become the fastest-growing factor in HCC in some western countries [4]. The leading factor contributing to the development of NAFLD is overeating, which leads to the accumulation of adipose cells in the liver tissue. NAFLD is interrelated with metabolic syndromes (MS) such as insulin resistance (IR), obesity, and type 2 diabetes (T2DM), which increases the risk of cirrhosis and related complications [2]. Furthermore, studies have indicated that NAFLD is also related to cardiovascular diseases [5], chronic kidney diseases [6], and the long-term risk of developing extrahepatic cancers [7].

The pathogenesis of NAFLD is complex and not yet clearly understood, so there is still a lack of specific drugs for its treatment, and lifestyle interventions are currently being used but with limited success [1]. Studies suggest that the novel hypoglycemic drug class of sodium-glucose cotransporter-2 (SGLT-2) inhibitors shows a favorable effect in the treatment of NAFLD [8, 9]. Thus, this review summarizes the clinical studies and possible mechanisms of SGLT-2 inhibitors in the treatment of NAFLD and may provide some ideas for the treatment of NAFLD.

2. The Pathogenesis of NAFLD

The pathogenesis of NAFLD is complicated and still needs to be further investigated. The “multiple-hit” hypothesis proposed that multiple injuries act together on genetically susceptible subjects to induce NAFLD. These injuries include adipokines secreted by adipose tissue, IR, gut microbiota, and inflammation [10] (Fig. 1).

Fig. 1.

Pathogenesis of NAFLD. DNL, de novo lipogenesis.

The main driver of NAFLD is overnutrition, which leads to the excessive accumulation of adipose tissue in the liver and exceeds the liver’s metabolic capacity [2]. Accumulation of high levels of free fatty acids (FFA), free cholesterol, and other lipid metabolites leads to increased lipotoxicity [10], further contributing to cellular stress such as oxidative stress and endoplasmic reticulum (ER) stress [2], production of reactive oxygen species (ROS), mitochondrial dysfunction [10, 11], inflammasome activation [12], and apoptotic cell death, and subsequent inflammatory stimulation, tissue regeneration, and fibrogenesis [2].

Adipokines secreted by adipose tissue in the liver promote the development of NAFLD. (1) Leptin is synthesized and secreted from fat cells [13]. Higher levels of circulating leptin are associated with increased severity of NAFLD can boost inflammation and can increase susceptibility to hepatotoxicity [14]. (2) Adiponectin is a kind of adipokine that possesses hepatoprotective effects. Decreased levels of adiponectin have been found in individuals with NAFLD, and the levels of adiponectin are inversely related to the severity of steatosis, necroinflammation, and fibrosis [14].

IR, which is characterized by reduced glucose disposal in adipose tissue or muscle, is a crucial factor in the development of NAFLD [10, 15]. Research has suggested that IR drives hepatic de novo lipogenesis (DNL) in patients with NAFLD [16]. Hepatic DNL is a highly regulated metabolic pathway by which cells convert excess carbohydrates (usually glucose) into fatty acids [11], and then cause the inappropriate release of fatty acids through dysregulated lipolysis, subsequently leading to impaired systemic insulin signaling [15].

Growing evidence has suggested that gut microbiota also acts as a significant driver to facilitate the occurrence and development of NAFLD, and to accelerate its progression to cirrhosis and HCC. The gut microbiota and its metabolites can directly affect intestinal morphology and immune response, leading to abnormal activation of inflammation and intestinal endotoxemia. Dysbiosis of the gut microbiota can also result in dysfunction of the gut-liver axis (physiological crosstalk between the liver and gut [12]) by changing the bile-acid metabolic pathway [17, 18].

Inflammatory activation is involved in the occurrence of NAFLD/NASH. Inflammation of the liver triggered by hepatocyte death, gut barrier dysfunction, adipokines, etc., contribute to the development of NAFLD [19]; the development of NASH is also the result of abnormal activation of conventional immune cells, parenchymal cells and endothelial cells of the liver in response to inflammatory mediators from the liver, adipose tissue and the gut [20].

3. The Link between NAFLD and T2DM

As mentioned above, NAFLD has a bidirectional association with IR, obesity, and T2DM [2]. NAFLD and T2DM are two pathological conditions that often coexist and interact with each other, and T2DM can increase the risk of adverse hepatic and extra-hepatic outcomes [21]. A large meta-analysis involving 49,419 T2DM patients from 20 countries demonstrated that the global prevalence of NAFLD in T2DM patients is 55.5% [22]. There is growing evidence indicating that NAFLD may precede or promote the development of T2DM and that the risk of developing T2DM parallels the severity of NAFLD [21]. Mechanically, evidence suggests that hepatic lipid accumulation is associated with IR and an increased risk of T2DM, and reduced hepatic lipid accumulation is also associated with a reduced risk of developing T2DM [21, 23]. However, the specific mechanism of the increased risk of T2DM in NAFLD patients has not been clarified.

4. Treatment for NAFLD
4.1 Pharmacotherapy
4.1.1 Hypoglycemic Drugs

Since NAFLD is tightly related to T2DM, and IR also plays a vital role in the pathogenesis of NAFLD, hypoglycemic drugs have shown beneficial effects in the treatment of NAFLD (Table 1, Ref. [24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42]). Among these glucose-lowering drugs, SGLT-2 inhibitors have promising applications due to their advantages of improving intrahepatic IR and alleviating hepatic steatosis, etc.

Table 1.Hypoglycemic drugs in the treatment of NAFLD.
Hypoglycemic drugs Hepatic beneficial effects Adverse effects
SGLT-2 inhibitors steatosis [24] and liver fat [29] risk for diabetic ketoacidosis [30]
ALT, GGT, LFC [25] mycotic genital infections [31]
weight [26] UTIs [31]
triglyceride, HDL-c [27]
IR, insulin sensitivity [28]
Pioglitazone NASH resolution [32] weight [32]
TG [32] CHF [35]
HOMA-IR [33] peripheral edema [35]
AST, ALT [34] bone fracture [35]
GLP-1RA insulin sensitivity [36] gastric mobility [36]
lipotoxicity and liver fat deposition [36] vomiting and diarrhea [39]
steatosis, ballooning, and lobular infiltrates of NASH [37]
weight [38]
DPP-IV inhibitors NAS and fibrosis [40] hospitalisation for heart failure [42]
lipogenesis [41]

Note: , decrease; , increase; GLP-1RA, Glucagon-Like Peptide Receptor Agonist; DPP-Ⅳ, Dipeptidyl Peptidase-IV; ALT, alanine aminotransferase; AST, aspartate aminotransferase; GGT, gamma-glutamyl transferase; HOMA-IR, Homoeostasis Model Assessment of Insulin Resistance; NAS, NAFLD activity score; LFC, liver fat content; TG, triglyceride; HDL-c, HDL cholesterol; CHF, congestive heart failure; SGLT-2, sodium-glucose co-transporter type 2; NASH, non-alcoholic steatohepatitis; UTIs, urinary tract infections.

4.1.1.1 Sodium-Glucose Co-Transporter Type 2 (SGLT-2) Inhibitors

SGLT-2 inhibitors (e.g., canagliflozin, dapagliflozin, empagliflozin) are the newest class of anti-diabetic medications and have shown some beneficial effects in the treatment of NAFLD. Clinical trials suggest that SGLT-2 inhibitors treatment is associated with the improvement of liver steatosis in NAFLD [24]. A meta-analysis shows that SGLT-2 inhibitors are effective in reducing aspartate aminotransferase (ALT), gamma-glutamyl transferase (GGT), and can also decrease the absolute percentage of liver fat content (LFC) on MRI in overweight or obese patients with NAFLD [25]. SGLT-2 inhibitors can reduce body weight in patients with or without T2DM [26, 43, 44], decrease plasma triglyceride (TG), and increase high-density lipoprotein (HDL) to improve dyslipidemia [27], which can be profoundly helpful in alleviating hepatic steatosis and improving NAFLD [45, 46]. SGLT-2 inhibitors can also alleviate IR and improve insulin sensitivity in high-fat diet (HFD) mice [28]. Furthermore, animal studies suggest evidence that canagliflozin attenuated the development of NASH and prevented the progression of NASH to HCC in the mouse model of diabetes and NASH/HCC [47]. In summary, SGLT-2 inhibitors can reduce LFC, improve hepatic steatosis, lower liver enzyme levels, improve dyslipidemia in patients with NAFLD, as well as improve IR, enhance insulin sensitivity and prevent the progression of NASH to HCC in mouse models, and SGLT-2 inhibitors probably are among the potentially effective drugs for the treatment of NAFLD.

4.1.1.2 Pioglitazone

As an insulin sensitizer, pioglitazone has been shown to improve liver function in NAFLD or NASH subjects [48]. Clinical study results showed that pioglitazone treatment improved fibrosis scores, reduced liver TG levels, improved insulin sensitivity in the liver, and made the resolution of NASH [32]. Treatment with pioglitazone, even at low doses, can also significantly improve hepatic steatosis in T2DM patients [33].

4.1.1.3 Glucagon-Like Peptide Receptor Agonists (GLP-1RAs)

GLP-1RAs (e.g., dulaglutide, exenatide, semaglutide) can reduce lipotoxicity by decreasing liver fat deposition, increasing hepatic insulin sensitivity by reducing steatosis, modulating liver fibrosis, and slowing down the progression of NAFLD to NASH [36]. GLP1-RAs can ameliorate steatosis, ballooning, and lobular infiltrates of NASH or achieve remission of NASH without worsening fibrosis [37].

4.1.1.4 Dipeptidyl Peptidase (DPP)-IV Inhibitors

DPP-IV inhibitors (e.g., sitagliptin, saxagliptin, linagliptin) are agents that can improve IR. Experiment have revealed that DPP-IV inhibitors attenuate NAFLD activity score (NAS) and fibrosis stage in the HFD-induced NAFLD mouse model [40]. The beneficial effect was likely achieved by restoring the gut-liver axis [49], reducing hepatic lipogenesis, and downregulating the expression of adipogenesis genes [41]. However, the beneficial effects of DPP-IV inhibitors on human NAFLD are still debated in clinical trials [50, 51, 52, 53], and strong clinical evidence for their clinical use in NAFLD/NASH is still lacking [38].

Altogether, SGLT-2 inhibitors benefit NAFLD remission not only by directly improving intrahepatic steatosis, reducing LFC, and improving liver enzyme levels, but also by improving systemic metabolic status by reducing body weight, lowering plasma TG levels, improving IR, and increasing insulin sensitivity. Therefore, SGLT-2 inhibitors may be at the top of list of hypoglycemic agents in the treatment of NAFLD.

4.1.2 Novel Drugs for NAFLD

In addition to the above-mentioned hypoglycemic drugs that are effective in treating NAFLD, there are also several novel drugs are currently in development or have entered clinical trials, Table 2 (Ref. [54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81]) provides a brief overview of the experimental drugs that have progressed to clinical trials.

Table 2.Novel drugs for NAFLD that progressed to clinical trials.
Types of drugs Hepatic outcomes Diabetic outcomes Refs
FXR steatosis and fibrosis, NAS, ALT and AST weight, HDL-c, insulin and HOMA-IR, TC and LDL-c [54, 55, 56]
PPAR agonists GGT and ALT, LFC, insulin sensitivity TG and LDL-c, insulin sensitivity, β-cell function [57, 58, 59, 60]
THRβ agonists lipid content, fibrosis, NAS, ALT, AST and GGT LDL-c and TG [61, 62]
ACC inhibitors steatosis and fibrosis, lipid content, DNL, ALT TG [63, 64, 65, 66]
FAS inhibitors liver fat, DNL, ALT and AST LDL-c and TC, insulin sensitivity [67, 68]
SCD1 inhibitors LFC, NFS, ALT, AST and GGT HbA1c [69, 70]
DGAT inhibitors LFC, ALT and AST TG [71, 72, 73]
KHK inhibitors WLF, GGT insulin concentration, HOMA-IR [74]
MPC inhibitors steatosis, NAS, ALT, AST and GGT fasting insulin, HbA1c, insulin sensitivity, weight HOMA-IR [75, 76]
FGF19 fibrosis, LFC, NAS, ALT and AST TG, LDL-c [77, 78]
FGF21 fibrosis, HFF, ALT, AST and GGT TG, HDL-c, insulin sensitivity [79, 80, 81]

The effect of Semaglutide in Subjects With Non-cirrhotic Non-alcoholic Steatohepatitis (NCT04822181): A clinical study that is under Phase 3 study about the effect of Semaglutide, a GLP-1 receptor agonist, on NASH.

Note: , decrease; , increase; FXR, Farnesoid X Receptor; PPAR, peroxisome proliferator-activated receptor; THRβ, Thyroid Hormone Receptorβ; ACC, Acetyl-coenzyme A carboxylase; FAS, Fatty acid synthase; SCD1, Stearoyl-CoA desaturase 1; DGAT, diacylglycerol acyltransferase; KHK, Ketohexokinase; MPC, mitochondrial pyruvate carrier; FGF, Fibroblast growth factor; TC, total serum cholesterol; LDL-c, LDL cholesterol; LFC, Liver fat content; NFS, NAFLD fibrosis score; WLF, whole liver fat; HFF, hepatic fat fraction; HbA1c, hemoglobin A1c.

Due to the different groupings and drug doses in clinical trials, the p-values for the above data changes were not necessarily <0.05 in all groupings.

4.2 Lifestyle Interventions

Lifestyle interventions include a combination of calorie restriction and exercise [11], which are currently the therapeutic strategies for NAFLD in the absence of approved pharmacological therapies [82]. In patients with NAFLD, exercise alone without dietary intervention can reduce intrahepatic triglycerides (IHTG). A combination of diet and exercise can significantly reduce the level of IHTG over exercise alone, as well as ameliorate glucose control and insulin sensitivity [83]. Strong evidence showed that weight loss can alleviate liver disease to some extent;weight loss of 10% relieves NASH and improves liver fibrosis, whereas modest weight loss (>5%) can also have important benefits for NAFLD [84]. However, very few patients can achieve this goal through intensive lifestyle modifications [85]. Thus, it might be difficult to implement such strict lifestyle interventions in clinical practice.

4.3 Bariatric Surgery

Bariatric surgery is an effective treatment for weight loss and can lead to improvement in comorbidities as well [86]. A prospective study involving 109 patients found that after 1 year of follow-up, bariatric surgery contributed to the disappearance of NASH in nearly 85% of patients and the subsiding of fibrosis in 33% of patients [87]. Another 5-year follow-up of NASH patients who underwent bariatric surgery, showed results that were also consistent [88]. In addition, in patients with NASH and obesity, bariatric surgery is associated with a significantly lower risk of major adverse liver outcomes and major adverse cardiovascular events than did non-surgical treatment [89]. Bariatric surgery seems to be a beneficial option for obese patients with NASH, but the safety of bariatric surgery has not been well established [86]. Bariatric surgery may only be considered in highly selected patients with obesity and compensated cirrhosis [90].

5. Clinical Studies of SGLT-2 Inhibitors on NAFLD

The main common clinical SGLT-2 inhibitors include canagliflozin, empagliflozin, and dapagliflozin, and Table 3 (Ref. [29, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100]) shows a summary of clinical studies on different SGLT-2 inhibitors for the treatment of NAFLD.

Table 3.Clinical studies of SGLT-2 inhibitors in the treatment of NAFLD.
Types of drugs Study population Duration (weeks) Hepatic benefits Diabetic benefits Refs
Canagliflozin T2DM, NAFLD 48 HFF, ALT, AST and GGT HbA1c, BMI and FM [91]
T2DM, NAFLD 24 steatosis HbA1c, weight, HOMA-IR [92]
T2DM, NASH 12 fibrosis, AST, GGT HbA1c, BMI [93]
Dapagliflozin T2DM, NAFLD 24 ALT, GGT HbA1c, weight, HOMA-IR, VAT [94]
T2DM, NAFLD 12 ALT, AST and GGT HbA1c, weight, HOMA-IR, VAT [95]
T2DM, NAFLD 12 lipid contents, ALT HbA1c, weight, body fat [96]
NAFLD 12 ALT and AST HbA1c, weight, BMI [97]
Empagliflozin T2DM, NAFLD 20 liver fat, ALT [29]
NAFLD 24 steatosis, ALT and AST, LSM fasting insulin [98]
T2DM, NAFLD 24 steatosis and fibrosis, AST, LSM HbA1c [99]
T2DM, NASH 24 steatosis, fibrosis and ballooning, GGT BMI, FBG, TC [100]

Note: , decrease; , increase; , No difference compared to the control group; BMI, body mass index; FM, fat mass; VAT, visceral adipose tissue ; LSM, liver stiffness measurement; T2DM, type 2 diabetes; FBG, fasting blood glucose.

6. Mechanism of SGLT-2 Inhibitors in the Treatment of NAFLD

As previously mentioned, SGLT-2 inhibitors might be a potential approach for the treatment of NAFLD and have shown different beneficial effects. However, the exact mechanism by which SGLT-2 inhibitors ameliorate the liver injury associated with NAFLD remains unclear [101] (Fig. 2).

Fig. 2.

Overview of the mechanism of improvement of NAFLD by SGLT-2 inhibitors. TG, triglyceride; TC, total serum cholesterol; FFA, free fatty acid; IR, insulin resistance; DNL, de novo lipogenesis.

6.1 Increase Endogenous Glucose and Expedite Lipolysis

The liver is an important metabolic organ in which metabolic function is controlled by insulin and other metabolic hormones; aberrant energy metabolism in the liver is related to NAFLD [102]. Insulin is the main regulator of glucose, lipid, and protein metabolism. When plasma glucose concentration increases, insulin secretion of β cells is stimulated, and hyperinsulinemia and hyperglycemia act synergistically to inhibit endogenous glucose production (EGP) and lipolysis. Some research indicates that SGLT-2 inhibition increases EGP due to increased gluconeogenesis, but not glycogenolysis [103, 104].

Fat accumulation is the main cause of the occurrence and development of NAFLD; SGLT-2 inhibitors can inhibit the production of liver fat and improve hepatic lipid metabolic disorder.

In HFD-induced mouse models, administration of canagliflozin reduced hepatic lipid accumulation, downregulated lipogenesis markers (SREBP-1c and FASN), and upregulated lipolysis markers (CPT1a, ACOX1, and ACADM) [105]. Animal studies also suggest that canagliflozin can promote lipolysis by upregulating the hepatic zinc-α2-glycoprotein (ZAG) levels and further activating extracellular signal-regulated kinase (ERK) [106, 107].

Empagliflozin hinders hepatic lipogenesis by increasing the expression of PPAR-α (a nuclear receptor protein that promotes fatty acid β-oxidation and enhances lipid catabolism) and by decreasing the expression of lipogenic genes PPAR-γ, SREBP1c and FAS [28]. Studies indicate that empagliflozin increases the expression of SIRT-1 (a metabolic sensor that is involved in the regulation of lipid and carbohydrate metabolism in hepatocytes [108, 109]) to accelerate fatty acid oxidation [107]. The activation of the SIRT-1/PGC-α/PPAR-α pathway by SGLT-2 inhibitors promotes fatty acid oxidation [107]. Therefore, SGLT-2 inhibitor may play a role in the treatment of NAFLD by increasing endogenous glucose and increasing lipolysis.

6.2 Inhibit Inflammation to Improve Hepatic Steatosis and Fibrosis

Inflammation activation participates in the development of NAFLD and NASH; studies suggest that SGLT-2 inhibitors also can inhibit the process of inflammation and thereby alleviate liver steatosis and fibrosis.

Canagliflozin suppressed the production of inflammatory cytokines, such as TNF-α, MCP-1, IL-1β, and IL-6 to ameliorate NAFLD in mouse experiments [105]. Similarly, ipragliflozin [107] and empagliflozin [110, 111] improve hepatic steatosis by inhibiting production of inflammatory cytokines IL-1β, IL-6, IL-8, TNF-α, and MCP-1. Furthermore, empagliflozin ameliorates liver inflammation and liver fibrosis by attenuating the activation of the NLRP3 inflammasome in rat liver, and slows the progression of NAFLD to NASH [112, 113].

Dapagliflozin can reduce hepatic steatosis and fibrosis in high-carbohydrate high-fat-induced NAFLD [107], which is accomplished by decreasing the DNL enzyme ACC1 and upregulating the fatty acid oxidation enzyme ACOX1 [114]. It also significantly reduces inflammatory factors such as TNF-α, IL-1β and IL-18 in the liver homogenate to attenuate inflammation [115].

6.3 Inhibit Cellular Stress, Prevent Hepatocytes Apoptosis, and Ameliorate Liver Injury

Cellular stress, such as oxidative stress and ER stress, is an important factor that produces the death of hepatocytes. SGLT-2 inhibitors can alleviate cellular stress and prevent hepatocyte apoptosis.

Animal studies have suggested that canagliflozin reduces hepatic oxidative stress and enhances the activity of antioxidant enzymes as well as enhances total antioxidant capacity [106]. Canagliflozin can lower hepatic inflammatory cytokiness levels, decrease serum caspase-3 levels, and enhance the expression of hepatic Bcl-2 to prevent hepatocyte apoptosis [106, 116] (Caspase-3 and Bcl-2 are factors that belong to the liver apoptosis pathway and relate to the attenuation of liver injury [117]).

Empagliflozin can reduce HFD-induced ER stress by downregulating the expression of genes involved in ER stress, such as CHOP, ATF4, and Gadd45 [28], thereby inhibiting the apoptotic process in hepatocytes [118]. Additionally, empagliflozin can enhance autophagy of hepatic macrophages via the AMPK/mTOR signaling pathway, and further inhibit IL-17/IL-23 axis-mediated inflammatory responses, thereby significantly attenuating NAFLD-related liver injury [101].

Ipragliflozin can enhance the concentration of FGF21, a key hepatokine with beneficial properties [119], thus significantly improving oxidative stress, inflammation, and fibrosis [107].

6.4 Increase Hepatic Insulin Clearance, Regulate Hepatic Fatty Acid Metabolism and Insulin Function

Insulin is produced in islet cells of the pancreas and mostly cleared in the liver. Hepatic insulin clearance (HIC) helps regulate insulin action by controlling insulin availability in peripheral tissues, and is closely related to hepatic lipid content and glucose metabolism [120]. Reduced HIC, due to obesity and liver fat accumulation, is one of the causes of inappropriate hyperinsulinemia and IR [121], and hyperinsulinemia is thought to play an important role in the pathogenesis of NAFLD and NASH [122]. SGLT-2 inhibitors are believed to increase HIC and improve β-cell function [123], thus improving NAFLD and NASH. SGLT-2 inhibitors are related to increased HIC by reducing plasma TG concentration and elevating plasma β-hydroxybutyrate (BHB, is the most abundant ketone body in mammals; it is synthesized in the liver from fatty acids [124]), and manifested as the reduction of intrahepatic TG content and improvement of fatty liver [121].

7. The Treatment of SGLT-2 Inhibitors in Comorbidities of NAFLD

The influence of NAFLD is not limited to the liver. NAFLD has extra-hepatic consequences such as renal and cardiovascular disease. Chronic kidney disease (CKD) and NAFLD share pathogenic mechanisms including IR, lipotoxicity, inflammation, and oxidative stress [125]. SGLT-2 inhibitors are sodium-glucose cotransporter inhibitors, which have hypoglycemic effects by inhibiting the reabsorption of glucose in the proximal convoluted tubule, leading to substantial glucosuria and a reduction in plasma glucose levels. SGLT-2 inhibitors reduce afferent arteriolar vasoconstriction and may provide nephroprotection by attenuating glomerular hyperfiltration [126], decreasing inflammation, fibrogenic responses and cell apoptosis, preventing glucose overload-induced oxidative stress [127] and reducing serum uric acid levels [128]. SGLT-2 inhibitors also have the potential for cardiovascular protection by reducing inflammation [129], expediting lipolysis, and by other mechanisms.

8. Adverse Effects for SGLT-2 Inhibitors and Potential Limitations in Clinical Settings

Although the SGLT-2 inhibitors play a crucial role in the treatment of NAFLD, there are adverse effects and potential limitations in their clinical use. First, SGLT-2 inhibitors can significantly increase the risk of urinary tract and genital infections [130]. Therefore, clinicians should avoid adding SGLT-2 inhibitors in people with pre-existing urinary tract infections. Patients should also be advised to increase water intake and for women to keep the vulva clean during treatment. Second, SGLT-2 inhibitors reportedly can increase the risk of diabetic ketoacidosis. In some cases, blood glucose levels are normal or only mildly elevated, which may delay diagnosis. Hence, clinicians should be aware of identifying the presence of diabetic ketoacidosis. Regardless of blood glucose levels, patients taking SGLT-2 inhibitors who have symptoms of, or triggers for, ketoacidosis should be checked for ketone levels [30]. Third, T2DM is a risk factor for osteoporosis, and previous studies have suggested that the use of SGLT-2 inhibitors may increase the risk of fractures. However, recent studies have suggested otherwise [131], and the effect of SGLT-2 inhibitors on fractures is still controversial. Therefore, bone mineral density and fracture-risk assessment should be considered in the clinical setting for patients at higher risk of fracture [132]. Fourth, recent studies suggest that there is no evidence of increased renal risks in using SGLT-2 inhibitors; canagliflozin [133] and dapagliflozin [134, 135] are approved for chronic kidney disease and diabetic kidney disease (DKD); also, the annual rate of decline in the estimated glomerular filtration rate(eGFR) was slower in the empagliflozin group than in the placebo group [136]. Consequently, SGLT-2 inhibitors can be considered for treatment of stage 4 CKD and albuminuria.

9. Conclusions

Taken together, the results suggest that SGLT-2 inhibitors show potential for the beneficial treatment of NAFLD by alleviating hepatic steatosis and fibrosis, lowering liver enzyme levels, improving hepatic and systemic insulin resistance, reducing body weight, and so on. In patients with NAFLD combined with T2DM, SGLT-2 inhibitors are recommended in the absence of contraindications. For patients with NAFLD combined with obesity, SGLT-2 inhibitors have also shown some therapeutic effects, and if weight loss is difficult to achieve or ineffective, SGLT-2 inhibitors can be tried without contraindications. However, the application of SGLT-2 inhibitors requires attention to possible side effects such asdiabetic ketoacidosis, and urogenital infection, and monitoring of liver and kidney function is highly recommended.

Abbreviations

IR, insulin resistance; TG, triglyceride; NAS, NAFLD activity score; LFC, liver fat content; HDL, high-density lipoprotein; HFD, high-fat diet; PPAR-α, peroxisome proliferator-activated receptor alpha; SREBP1c, sterol regulatory element-binding transcription factor 1; FAS/FASN, fatty acid synthase; Gadd45, growth arrest and DNA damage-inducible protein; ApoE, Apolipoprotein E; TNF-α,Tumor necrosis factor-alpha; IL, interleukin; ACC1, acetyl-CoA carboxylase 1; ACOX1, acyl-CoA oxidase 1; ER, endoplasmic reticulum; Caspase-3, cysteinyl aspartate specific proteinase-3; MCP-1, Monocyte Chemoattractant Protein-1; FGF, fibroblast growth factor; MS, metabolic syndrome; HIC, hepatic insulin clearance; FFA, free fatty acids; eGFR, estimated glomerular filtration rate.

Author Contributions

BZ provided the conception and design of the manuscript, and agree to be accountable for all aspects of the work; RX and DL drafted the manuscript and reviewed the intellectual content critically; ZC and YX analyzed and interpreted data of the work; YanW, LM and YuanW acquired data of the work, and reviewed the manuscript critically. All the authors read and approved the manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Fu Y, Zhou Y, Shen L, Li X, Zhang H, Cui Y, et al. Diagnostic and therapeutic strategies for non-alcoholic fatty liver disease. Frontiers in Pharmacology. 2022; 13: 973366.
[2]
Powell EE, Wong VWS, Rinella M. Non-alcoholic fatty liver disease. Lancet. 2021; 397: 2212–2224.
[3]
Maurice J, Manousou P. Non-alcoholic fatty liver disease. Clinical Medicine. 2018; 18: 245–250.
[4]
Huang DQ, El-Serag HB, Loomba R. Global epidemiology of NAFLD-related HCC: trends, predictions, risk factors and prevention. Nature Reviews. Gastroenterology & Hepatology. 2021; 18: 223–238.
[5]
Polyzos SA, Kechagias S, Tsochatzis EA. Review article: non-alcoholic fatty liver disease and cardiovascular diseases: associations and treatment considerations. Alimentary Pharmacology & Therapeutics. 2021; 54: 1013–1025.
[6]
Adams LA, Anstee QM, Tilg H, Targher G. Non-alcoholic fatty liver disease and its relationship with cardiovascular disease and other extrahepatic diseases. Gut. 2017; 66: 1138–1153.
[7]
Mantovani A, Petracca G, Beatrice G, Csermely A, Tilg H, Byrne CD, et al. Non-alcoholic fatty liver disease and increased risk of incident extrahepatic cancers: a meta-analysis of observational cohort studies. Gut. 2022; 71: 778–788.
[8]
Bellanti F, Lo Buglio A, Dobrakowski M, Kasperczyk A, Kasperczyk S, Aich P, et al. Impact of sodium glucose cotransporter-2 inhibitors on liver steatosis/fibrosis/inflammation and redox balance in non-alcoholic fatty liver disease. World Journal of Gastroenterology. 2022; 28: 3243–3257.
[9]
Arai T, Atsukawa M, Tsubota A, Mikami S, Haruki U, Yoshikata K, et al. Antifibrotic effect and long-term outcome of SGLT2 inhibitors in patients with NAFLD complicated by diabetes mellitus. Hepatology Communications. 2022; 6: 3073–3082.
[10]
Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016; 65: 1038–1048.
[11]
Nassir F. NAFLD: Mechanisms, Treatments, and Biomarkers. Biomolecules. 2022; 12: 824.
[12]
Schuster S, Cabrera D, Arrese M, Feldstein AE. Triggering and resolution of inflammation in NASH. Nature Reviews. Gastroenterology & Hepatology. 2018; 15: 349–364.
[13]
Obradovic M, Sudar-Milovanovic E, Soskic S, Essack M, Arya S, Stewart AJ, et al. Leptin and Obesity: Role and Clinical Implication. Frontiers in Endocrinology. 2021; 12: 585887.
[14]
Chang ML, Yang Z, Yang SS. Roles of Adipokines in Digestive Diseases: Markers of Inflammation, Metabolic Alteration and Disease Progression. International Journal of Molecular Sciences. 2020; 21: 8308.
[15]
Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nature Medicine. 2018; 24: 908–922.
[16]
Smith GI, Shankaran M, Yoshino M, Schweitzer GG, Chondronikola M, Beals JW, et al. Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease. The Journal of Clinical Investigation. 2020; 130: 1453–1460.
[17]
Fang J, Yu CH, Li XJ, Yao JM, Fang ZY, Yoon SH, et al. Gut dysbiosis in nonalcoholic fatty liver disease: pathogenesis, diagnosis, and therapeutic implications. Frontiers in Cellular and Infection Microbiology. 2022; 12: 997018.
[18]
Chen J, Vitetta L. Gut Microbiota Metabolites in NAFLD Pathogenesis and Therapeutic Implications. International Journal of Molecular Sciences. 2020; 21: 5214.
[19]
Wang H, Mehal W, Nagy LE, Rotman Y. Immunological mechanisms and therapeutic targets of fatty liver diseases. Cellular & Molecular Immunology. 2021; 18: 73–91.
[20]
Luci C, Bourinet M, Leclère PS, Anty R, Gual P. Chronic Inflammation in Non-Alcoholic Steatohepatitis: Molecular Mechanisms and Therapeutic Strategies. Frontiers in Endocrinology. 2020; 11: 597648.
[21]
Targher G, Corey KE, Byrne CD, Roden M. The complex link between NAFLD and type 2 diabetes mellitus - mechanisms and treatments. Nature Reviews. Gastroenterology & Hepatology. 2021; 18: 599–612.
[22]
Younossi ZM, Golabi P, de Avila L, Paik JM, Srishord M, Fukui N, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. Journal of Hepatology. 2019; 71: 793–801.
[23]
Sung KC, Wild SH, Byrne CD. Resolution of fatty liver and risk of incident diabetes. The Journal of Clinical Endocrinology and Metabolism. 2013; 98: 3637–3643.
[24]
Coelho FDS, Borges-Canha M, von Hafe M, Neves JS, Vale C, Leite AR, et al. Effects of sodium-glucose co-transporter 2 inhibitors on liver parameters and steatosis: A meta-analysis of randomized clinical trials. Diabetes/metabolism Research and Reviews. 2021; 37: e3413.
[25]
Mantovani A, Petracca G, Csermely A, Beatrice G, Targher G. Sodium-Glucose Cotransporter-2 Inhibitors for Treatment of Nonalcoholic Fatty Liver Disease: A Meta-Analysis of Randomized Controlled Trials. Metabolites. 2020; 11: 22.
[26]
Brown E, Heerspink HJL, Cuthbertson DJ, Wilding JPH. SGLT2 inhibitors and GLP-1 receptor agonists: established and emerging indications. Lancet. 2021; 398: 262–276.
[27]
Hayashi T, Fukui T, Nakanishi N, Yamamoto S, Tomoyasu M, Osamura A, et al. Dapagliflozin decreases small dense low-density lipoprotein-cholesterol and increases high-density lipoprotein 2-cholesterol in patients with type 2 diabetes: comparison with sitagliptin. Cardiovascular Diabetology. 2017; 16: 8.
[28]
Petito-da-Silva TI, Souza-Mello V, Barbosa-da-Silva S. Empaglifozin mitigates NAFLD in high-fat-fed mice by alleviating insulin resistance, lipogenesis and ER stress. Molecular and Cellular Endocrinology. 2019; 498: 110539.
[29]
Kuchay MS, Krishan S, Mishra SK, Farooqui KJ, Singh MK, Wasir JS, et al. Effect of Empagliflozin on Liver Fat in Patients With Type 2 Diabetes and Nonalcoholic Fatty Liver Disease: A Randomized Controlled Trial (E-LIFT Trial). Diabetes Care. 2018; 41: 1801–1808.
[30]
Musso G, Saba F, Cassader M, Gambino R. Diabetic ketoacidosis with SGLT2 inhibitors. British Medical Journal. 2020; 371: m4147.
[31]
Scheen AJ. Pharmacodynamics, efficacy and safety of sodium-glucose co-transporter type 2 (SGLT2) inhibitors for the treatment of type 2 diabetes mellitus. Drugs. 2015; 75: 33–59.
[32]
Cusi K, Orsak B, Bril F, Lomonaco R, Hecht J, Ortiz-Lopez C, et al. Long-Term Pioglitazone Treatment for Patients With Nonalcoholic Steatohepatitis and Prediabetes or Type 2 Diabetes Mellitus: A Randomized Trial. Annals of Internal Medicine. 2016; 165: 305–315.
[33]
Della Pepa G, Russo M, Vitale M, Carli F, Vetrani C, Masulli M, et al. Pioglitazone even at low dosage improves NAFLD in type 2 diabetes: clinical and pathophysiological insights from a subgroup of the TOSCA.IT randomised trial. Diabetes Research and Clinical Practice. 2021; 178: 108984.
[34]
Lian J, Fu J. Pioglitazone for NAFLD Patients With Prediabetes or Type 2 Diabetes Mellitus: A Meta-Analysis. Frontiers in Endocrinology. 2021; 12: 615409.
[35]
Shah P, Mudaliar S. Pioglitazone: side effect and safety profile. Expert Opinion on Drug Safety. 2010; 9: 347–354.
[36]
Laurindo LF, Barbalho SM, Guiguer EL, da Silva Soares de Souza M, de Souza GA, Fidalgo TM, et al. GLP-1a: Going beyond Traditional Use. International Journal of Molecular Sciences. 2022; 23: 739.
[37]
Mantovani A, Byrne CD, Targher G. Efficacy of peroxisome proliferator-activated receptor agonists, glucagon-like peptide-1 receptor agonists, or sodium-glucose cotransporter-2 inhibitors for treatment of non-alcoholic fatty liver disease: a systematic review. The Lancet. Gastroenterology & Hepatology. 2022; 7: 367–378.
[38]
Gastaldelli A, Cusi K. From NASH to diabetes and from diabetes to NASH: Mechanisms and treatment options. JHEP Reports: Innovation in Hepatology. 2019; 1: 312–328.
[39]
Nauck MA, Quast DR, Wefers J, Meier JJ. GLP-1 receptor agonists in the treatment of type 2 diabetes - state-of-the-art. Molecular Metabolism. 2021; 46: 101102.
[40]
Oh JH, Jun DW, Kim HY, Lee SM, Yoon EL, Hwang J, et al. Discovery of dipeptidyl peptidase-4 inhibitor specific biomarker in non-alcoholic fatty liver disease mouse models using modified basket trial. Clinical and Molecular Hepatology. 2022; 28: 497–509.
[41]
Ideta T, Shirakami Y, Miyazaki T, Kochi T, Sakai H, Moriwaki H, et al. The Dipeptidyl Peptidase-4 Inhibitor Teneligliptin Attenuates Hepatic Lipogenesis via AMPK Activation in Non-Alcoholic Fatty Liver Disease Model Mice. International Journal of Molecular Sciences. 2015; 16: 29207–29218.
[42]
Deacon CF, Lebovitz HE. Comparative review of dipeptidyl peptidase-4 inhibitors and sulphonylureas. Diabetes, Obesity & Metabolism. 2016; 18: 333–347.
[43]
Heerspink HJL, Perkins BA, Fitchett DH, Husain M, Cherney DZI. Sodium Glucose Cotransporter 2 Inhibitors in the Treatment of Diabetes Mellitus: Cardiovascular and Kidney Effects, Potential Mechanisms, and Clinical Applications. Circulation. 2016; 134: 752–772.
[44]
Wong J, Chan KY, Lo K. Sodium-glucose co-transporter 2 inhibitors on weight change and cardiometabolic profiles in individuals with overweight or obesity and without diabetes: A meta-analysis. Obesity Reviews. 2021; 22: e13336.
[45]
Koutoukidis DA, Astbury NM, Tudor KE, Morris E, Henry JA, Noreik M, et al. Association of Weight Loss Interventions With Changes in Biomarkers of Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-analysis. JAMA Internal Medicine. 2019; 179: 1262–1271.
[46]
Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: An update. Metabolism. 2016; 65: 1109–1123.
[47]
Jojima T, Wakamatsu S, Kase M, Iijima T, Maejima Y, Shimomura K, et al. The SGLT2 Inhibitor Canagliflozin Prevents Carcinogenesis in a Mouse Model of Diabetes and Non-Alcoholic Steatohepatitis-Related Hepatocarcinogenesis: Association with SGLT2 Expression in Hepatocellular Carcinoma. International Journal of Molecular Sciences. 2019; 20: 5237.
[48]
Bifari F, Manfrini R, Dei Cas M, Berra C, Siano M, Zuin M, et al. Multiple target tissue effects of GLP-1 analogues on non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Pharmacological Research. 2018; 137: 219–229.
[49]
Silva-Veiga FM, Miranda CS, Vasques-Monteiro IML, Souza-Tavares H, Martins FF, Daleprane JB, et al. Peroxisome proliferator-activated receptor-alpha activation and dipeptidyl peptidase-4 inhibition target dysbiosis to treat fatty liver in obese mice. World Journal of Gastroenterology. 2022; 28: 1814–1829.
[50]
Yilmaz Y, Yonal O, Deyneli O, Celikel CA, Kalayci C, Duman DG. Effects of sitagliptin in diabetic patients with nonalcoholic steatohepatitis. Acta Gastro-enterologica Belgica. 2012; 75: 240–244.
[51]
Arase Y, Kawamura Y, Seko Y, Kobayashi M, Suzuki F, Suzuki Y, et al. Efficacy and safety in sitagliptin therapy for diabetes complicated by non-alcoholic fatty liver disease. Hepatology Research. 2013; 43: 1163–1168.
[52]
Deng XL, Ma R, Zhu HX, Zhu J. Short article: A randomized-controlled study of sitagliptin for treating diabetes mellitus complicated by nonalcoholic fatty liver disease. European Journal of Gastroenterology & Hepatology. 2017; 29: 297–301.
[53]
Dos Santos LR, Duarte ML, Peccin MS, Gagliardi ARDT, Melnik T. Dipeptidyl Peptidase IV Inhibitors for Nonalcoholic Fatty Liver Disease - Systematic Review and Metanalysis. Current Diabetes Reviews. 2021; 17: e101120187811.
[54]
Neuschwander-Tetri BA, Loomba R, Sanyal AJ, Lavine JE, Van Natta ML, Abdelmalek MF, et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet. 2015; 385: 956–965.
[55]
Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, Goodman Z, et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019; 394: 2184–2196.
[56]
Patel K, Harrison SA, Elkhashab M, Trotter JF, Herring R, Rojter SE, et al. Cilofexor, a Nonsteroidal FXR Agonist, in Patients With Noncirrhotic NASH: A Phase 2 Randomized Controlled Trial. Hepatology. 2020; 72: 58–71.
[57]
Cariou B, Hanf R, Lambert-Porcheron S, Zaïr Y, Sauvinet V, Noël B, et al. Dual peroxisome proliferator-activated receptor α/δ agonist GFT505 improves hepatic and peripheral insulin sensitivity in abdominally obese subjects. Diabetes Care. 2013; 36: 2923–2930.
[58]
Jain N, Bhansali S, Kurpad AV, Hawkins M, Sharma A, Kaur S, et al. Effect of a Dual PPAR α/γ agonist on Insulin Sensitivity in Patients of Type 2 Diabetes with Hypertriglyceridemia- Randomized double-blind placebo-controlled trial. Scientific Reports. 2019; 9: 19017.
[59]
Gawrieh S, Noureddin M, Loo N, Mohseni R, Awasty V, Cusi K, et al. Saroglitazar, a PPAR-α/γ Agonist, for Treatment of NAFLD: A Randomized Controlled Double-Blind Phase 2 Trial. Hepatology. 2021; 74: 1809–1824.
[60]
Francque SM, Bedossa P, Ratziu V, Anstee QM, Bugianesi E, Sanyal AJ, et al. A Randomized, Controlled Trial of the Pan-PPAR Agonist Lanifibranor in NASH. The New England Journal of Medicine. 2021; 385: 1547–1558.
[61]
Harrison SA, Bashir MR, Guy CD, Zhou R, Moylan CA, Frias JP, et al. Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2019; 394: 2012–2024.
[62]
Taub R, Chiang E, Chabot-Blanchet M, Kelly MJ, Reeves RA, Guertin MC, et al. Lipid lowering in healthy volunteers treated with multiple doses of MGL-3196, a liver-targeted thyroid hormone receptor-β agonist. Atherosclerosis. 2013; 230: 373–380.
[63]
Kim CW, Addy C, Kusunoki J, Anderson NN, Deja S, Fu X, et al. Acetyl CoA Carboxylase Inhibition Reduces Hepatic Steatosis but Elevates Plasma Triglycerides in Mice and Humans: A Bedside to Bench Investigation. Cell Metabolism. 2017; 26: 394–406.e6.
[64]
Loomba R, Kayali Z, Noureddin M, Ruane P, Lawitz EJ, Bennett M, et al. GS-0976 Reduces Hepatic Steatosis and Fibrosis Markers in Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology. 2018; 155: 1463–1473.e6.
[65]
Lawitz EJ, Coste A, Poordad F, Alkhouri N, Loo N, McColgan BJ, et al. Acetyl-CoA Carboxylase Inhibitor GS-0976 for 12 Weeks Reduces Hepatic De Novo Lipogenesis and Steatosis in Patients With Nonalcoholic Steatohepatitis. Clinical Gastroenterology and Hepatology. 2018; 16: 1983–1991.e3.
[66]
Alkhouri N, Lawitz E, Noureddin M, DeFronzo R, Shulman GI. GS-0976 (Firsocostat): an investigational liver-directed acetyl-CoA carboxylase (ACC) inhibitor for the treatment of non-alcoholic steatohepatitis (NASH). Expert Opinion on Investigational Drugs. 2020; 29: 135–141.
[67]
Syed-Abdul MM, Parks EJ, Gaballah AH, Bingham K, Hammoud GM, Kemble G, et al. Fatty Acid Synthase Inhibitor TVB-2640 Reduces Hepatic de Novo Lipogenesis in Males With Metabolic Abnormalities. Hepatology. 2020; 72: 103–118.
[68]
Loomba R, Mohseni R, Lucas KJ, Gutierrez JA, Perry RG, Trotter JF, et al. TVB-2640 (FASN Inhibitor) for the Treatment of Nonalcoholic Steatohepatitis: FASCINATE-1, a Randomized, Placebo-Controlled Phase 2a Trial. Gastroenterology. 2021; 161: 1475–1486.
[69]
Safadi R, Konikoff FM, Mahamid M, Zelber-Sagi S, Halpern M, Gilat T, et al. The fatty acid-bile acid conjugate Aramchol reduces liver fat content in patients with nonalcoholic fatty liver disease. Clinical Gastroenterology and Hepatology. 2014; 12: 2085–2091.e1.
[70]
Ratziu V, de Guevara L, Safadi R, Poordad F, Fuster F, Flores-Figueroa J, et al. Aramchol in patients with nonalcoholic steatohepatitis: a randomized, double-blind, placebo-controlled phase 2b trial. Nature Medicine. 2021; 27: 1825–1835.
[71]
Loomba R, Morgan E, Watts L, Xia S, Hannan LA, Geary RS, et al. Novel antisense inhibition of diacylglycerol O-acyltransferase 2 for treatment of non-alcoholic fatty liver disease: a multicentre, double-blind, randomised, placebo-controlled phase 2 trial. The Lancet. Gastroenterology & Hepatology. 2020; 5: 829–838.
[72]
Amin NB, Carvajal-Gonzalez S, Purkal J, Zhu T, Crowley C, Perez S, et al. Targeting diacylglycerol acyltransferase 2 for the treatment of nonalcoholic steatohepatitis. Science Translational Medicine. 2019; 11: eaav9701.
[73]
Denison H, Nilsson C, Löfgren L, Himmelmann A, Mårtensson G, Knutsson M, et al. Diacylglycerol acyltransferase 1 inhibition with AZD7687 alters lipid handling and hormone secretion in the gut with intolerable side effects: a randomized clinical trial. Diabetes, Obesity & Metabolism. 2014; 16: 334–343.
[74]
Kazierad DJ, Chidsey K, Somayaji VR, Bergman AJ, Birnbaum MJ, Calle RA. Inhibition of ketohexokinase in adults with NAFLD reduces liver fat and inflammatory markers: A randomized phase 2 trial. Med. 2021; 2: 800–813.e3.
[75]
Harrison SA, Alkhouri N, Davison BA, Sanyal A, Edwards C, Colca JR, et al. Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled phase IIb study. Journal of Hepatology. 2020; 72: 613–626.
[76]
Ferguson D, Finck BN. Emerging therapeutic approaches for the treatment of NAFLD and type 2 diabetes mellitus. Nature Reviews. Endocrinology. 2021; 17: 484–495.
[77]
Harrison SA, Rinella ME, Abdelmalek MF, Trotter JF, Paredes AH, Arnold HL, et al. NGM282 for treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2018; 391: 1174–1185.
[78]
Harrison SA, Rossi SJ, Paredes AH, Trotter JF, Bashir MR, Guy CD, et al. NGM282 Improves Liver Fibrosis and Histology in 12 Weeks in Patients With Nonalcoholic Steatohepatitis. Hepatology. 2020; 71: 1198–1212.
[79]
Charles ED, Neuschwander-Tetri BA, Pablo Frias J, Kundu S, Luo Y, Tirucherai GS, et al. Pegbelfermin (BMS-986036), PEGylated FGF21, in Patients with Obesity and Type 2 Diabetes: Results from a Randomized Phase 2 Study. Obesity. 2019; 27: 41–49.
[80]
Harrison SA, Ruane PJ, Freilich BL, Neff G, Patil R, Behling CA, et al. Efruxifermin in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a trial. Nature Medicine. 2021; 27: 1262–1271.
[81]
Sanyal A, Charles ED, Neuschwander-Tetri BA, Loomba R, Harrison SA, Abdelmalek MF, et al. Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet. 2019; 392: 2705–2717.
[82]
El-Agroudy NN, Kurzbach A, Rodionov RN, O’Sullivan J, Roden M, Birkenfeld AL, et al. Are Lifestyle Therapies Effective for NAFLD Treatment? Trends in Endocrinology and Metabolism. 2019; 30: 701–709.
[83]
Golabi P, Locklear CT, Austin P, Afdhal S, Byrns M, Gerber L, et al. Effectiveness of exercise in hepatic fat mobilization in non-alcoholic fatty liver disease: Systematic review. World Journal of Gastroenterology. 2016; 22: 6318–6327.
[84]
Romero-Gómez M, Zelber-Sagi S, Trenell M. Treatment of NAFLD with diet, physical activity and exercise. Journal of Hepatology. 2017; 67: 829–846.
[85]
Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, Torres-Gonzalez A, Gra-Oramas B, Gonzalez-Fabian L, et al. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology. 2015; 149: 367–378.e5.
[86]
Sheka AC, Adeyi O, Thompson J, Hameed B, Crawford PA, Ikramuddin S. Nonalcoholic Steatohepatitis: A Review. The Journal of the American Medical Association. 2020; 323: 1175–1183.
[87]
Lassailly G, Caiazzo R, Buob D, Pigeyre M, Verkindt H, Labreuche J, et al. Bariatric Surgery Reduces Features of Nonalcoholic Steatohepatitis in Morbidly Obese Patients. Gastroenterology. 2015; 149: 379–388; quiz e15–e16.
[88]
Lassailly G, Caiazzo R, Ntandja-Wandji LC, Gnemmi V, Baud G, Verkindt H, et al. Bariatric Surgery Provides Long-term Resolution of Nonalcoholic Steatohepatitis and Regression of Fibrosis. Gastroenterology. 2020; 159: 1290–1301.e5.
[89]
Aminian A, Al-Kurd A, Wilson R, Bena J, Fayazzadeh H, Singh T, et al. Association of Bariatric Surgery With Major Adverse Liver and Cardiovascular Outcomes in Patients With Biopsy-Proven Nonalcoholic Steatohepatitis. The Journal of the American Medical Association. 2021; 326: 2031–2042.
[90]
Khajeh E, Aminizadeh E, Eslami P, Ramouz A, Kulu Y, Billeter AT, et al. Outcomes of bariatric surgery in patients with obesity and compensated liver cirrhosis. Surgery for Obesity and Related Diseases. 2022; 18: 727–737.
[91]
Inoue M, Hayashi A, Taguchi T, Arai R, Sasaki S, Takano K, et al. Effects of canagliflozin on body composition and hepatic fat content in type 2 diabetes patients with non-alcoholic fatty liver disease. Journal of Diabetes Investigation. 2019; 10: 1004–1011.
[92]
Nishimiya N, Tajima K, Imajo K, Kameda A, Yoshida E, Togashi Y, et al. Effects of Canagliflozin on Hepatic Steatosis, Visceral Fat and Skeletal Muscle among Patients with Type 2 Diabetes and Non-alcoholic Fatty Liver Disease. Internal Medicine. 2021; 60: 3391–3399.
[93]
Seko Y, Nishikawa T, Umemura A, Yamaguchi K, Moriguchi M, Yasui K, et al. Efficacy and safety of canagliflozin in type 2 diabetes mellitus patients with biopsy-proven nonalcoholic steatohepatitis classified as stage 1-3 fibrosis. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy. 2018; 11: 835–843.
[94]
Shimizu M, Suzuki K, Kato K, Jojima T, Iijima T, Murohisa T, et al. Evaluation of the effects of dapagliflozin, a sodium-glucose co-transporter-2 inhibitor, on hepatic steatosis and fibrosis using transient elastography in patients with type 2 diabetes and non-alcoholic fatty liver disease. Diabetes, Obesity & Metabolism. 2019; 21: 285–292.
[95]
Eriksson JW, Lundkvist P, Jansson PA, Johansson L, Kvarnström M, Moris L, et al. Effects of dapagliflozin and n-3 carboxylic acids on non-alcoholic fatty liver disease in people with type 2 diabetes: a double-blind randomised placebo-controlled study. Diabetologia. 2018; 61: 1923–1934.
[96]
Phrueksotsai S, Pinyopornpanish K, Euathrongchit J, Leerapun A, Phrommintikul A, Buranapin S, et al. The effects of dapagliflozin on hepatic and visceral fat in type 2 diabetes patients with non-alcoholic fatty liver disease. Journal of Gastroenterology and Hepatology. 2021; 36: 2952–2959.
[97]
Tobita H, Yazaki T, Kataoka M, Kotani S, Oka A, Mishiro T, et al. Comparison of dapagliflozin and teneligliptin in nonalcoholic fatty liver disease patients without type 2 diabetes mellitus: a prospective randomized study. Journal of Clinical Biochemistry and Nutrition. 2021; 68: 173–180.
[98]
Taheri H, Malek M, Ismail-Beigi F, Zamani F, Sohrabi M, Reza Babaei M, et al. Effect of Empagliflozin on Liver Steatosis and Fibrosis in Patients With Non-Alcoholic Fatty Liver Disease Without Diabetes: A Randomized, Double-Blind, Placebo-Controlled Trial. Advances in Therapy. 2020; 37: 4697–4708.
[99]
Chehrehgosha H, Sohrabi MR, Ismail-Beigi F, Malek M, Reza Babaei M, Zamani F, et al. Empagliflozin Improves Liver Steatosis and Fibrosis in Patients with Non-Alcoholic Fatty Liver Disease and Type 2 Diabetes: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Diabetes Therapy. 2021; 12: 843–861.
[100]
Lai LL, Vethakkan SR, Nik Mustapha NR, Mahadeva S, Chan WK. Empagliflozin for the Treatment of Nonalcoholic Steatohepatitis in Patients with Type 2 Diabetes Mellitus. Digestive Diseases and Sciences. 2020; 65: 623–631.
[101]
Meng Z, Liu X, Li T, Fang T, Cheng Y, Han L, et al. The SGLT2 inhibitor empagliflozin negatively regulates IL-17/IL-23 axis-mediated inflammatory responses in T2DM with NAFLD via the AMPK/mTOR/autophagy pathway. International Immunopharmacology. 2021; 94: 107492.
[102]
Rui L. Energy metabolism in the liver. Comprehensive Physiology. 2014; 4: 177–197.
[103]
Wolf P, Fellinger P, Pfleger L, Beiglböck H, Krumpolec P, Barbieri C, et al. Gluconeogenesis, But Not Glycogenolysis, Contributes to the Increase in Endogenous Glucose Production by SGLT-2 Inhibition. Diabetes Care. 2021; 44: 541–548.
[104]
Alatrach M, Agyin C, Solis-Herrera C, Lavryneko O, Adams J, Gastaldelli A, et al. Dapagliflozin Impairs the Suppression of Endogenous Glucose Production in Type 2 Diabetes Following Oral Glucose. Diabetes Care. 2022; 45: 1372–1380.
[105]
Xu Z, Hu W, Wang B, Xu T, Wang J, Wei D. Canagliflozin Ameliorates Nonalcoholic Fatty Liver Disease by Regulating Lipid Metabolism and Inhibiting Inflammation through Induction of Autophagy. Yonsei Medical Journal. 2022; 63: 619–631.
[106]
Kabil SL, Mahmoud NM. Canagliflozin protects against non-alcoholic steatohepatitis in type-2 diabetic rats through zinc alpha-2 glycoprotein up-regulation. European Journal of Pharmacology. 2018; 828: 135–145.
[107]
Ala M. SGLT2 Inhibition for Cardiovascular Diseases, Chronic Kidney Disease, and NAFLD. Endocrinology. 2021; 162: bqab157.
[108]
Nassir F, Ibdah JA. Sirtuins and nonalcoholic fatty liver disease. World Journal of Gastroenterology. 2016; 22: 10084–10092.
[109]
Liou CJ, Lee YK, Ting NC, Chen YL, Shen SC, Wu SJ, et al. Protective Effects of Licochalcone A Ameliorates Obesity and Non-Alcoholic Fatty Liver Disease Via Promotion of the Sirt-1/AMPK Pathway in Mice Fed a High-Fat Diet. Cells. 2019; 8: 447.
[110]
Jojima T, Tomotsune T, Iijima T, Akimoto K, Suzuki K, Aso Y. Empagliflozin (an SGLT2 inhibitor), alone or in combination with linagliptin (a DPP-4 inhibitor), prevents steatohepatitis in a novel mouse model of non-alcoholic steatohepatitis and diabetes. Diabetology & Metabolic Syndrome. 2016; 8: 45.
[111]
Lv Q, Le L, Xiang J, Jiang B, Chen S, Xiao P. Liver Transcriptomic Reveals Novel Pathways of Empagliflozin Associated With Type 2 Diabetic Rats. Frontiers in Endocrinology. 2020; 11: 111.
[112]
Benetti E, Mastrocola R, Vitarelli G, Cutrin JC, Nigro D, Chiazza F, et al. Empagliflozin Protects against Diet-Induced NLRP-3 Inflammasome Activation and Lipid Accumulation. The Journal of Pharmacology and Experimental Therapeutics. 2016; 359: 45–53.
[113]
Wree A, McGeough MD, Peña CA, Schlattjan M, Li H, Inzaugarat ME, et al. NLRP3 inflammasome activation is required for fibrosis development in NAFLD. Journal of Molecular Medicine. 2014; 92: 1069–1082.
[114]
Li L, Li Q, Huang W, Han Y, Tan H, An M, et al. Dapagliflozin Alleviates Hepatic Steatosis by Restoring Autophagy via the AMPK-mTOR Pathway. Frontiers in Pharmacology. 2021; 12: 589273.
[115]
ElMahdy MK, Helal MG, Ebrahim TM. Potential anti-inflammatory effect of dapagliflozin in HCHF diet- induced fatty liver degeneration through inhibition of TNF-α, IL-1β, and IL-18 in rat liver. International Immunopharmacology. 2020; 86: 106730.
[116]
Thapaliya S, Wree A, Povero D, Inzaugarat ME, Berk M, Dixon L, et al. Caspase 3 inactivation protects against hepatic cell death and ameliorates fibrogenesis in a diet-induced NASH model. Digestive Diseases and Sciences. 2014; 59: 1197–1206.
[117]
Hu Q, Zhang W, Wu Z, Tian X, Xiang J, Li L, et al. Baicalin and the liver-gut system: Pharmacological bases explaining its therapeutic effects. Pharmacological Research. 2021; 165: 105444.
[118]
Nasiri-Ansari N, Nikolopoulou C, Papoutsi K, Kyrou I, Mantzoros CS, Kyriakopoulos G, et al. Empagliflozin Attenuates Non-Alcoholic Fatty Liver Disease (NAFLD) in High Fat Diet Fed ApoE(-/-) Mice by Activating Autophagy and Reducing ER Stress and Apoptosis. International Journal of Molecular Sciences. 2021; 22: 818.
[119]
Parlati L, Régnier M, Guillou H, Postic C. New targets for NAFLD. JHEP Reports. 2021; 3: 100346.
[120]
Bojsen-Møller KN, Lundsgaard AM, Madsbad S, Kiens B, Holst JJ. Hepatic Insulin Clearance in Regulation of Systemic Insulin Concentrations-Role of Carbohydrate and Energy Availability. Diabetes. 2018; 67: 2129–2136.
[121]
Matsubayashi Y, Yoshida A, Suganami H, Osawa T, Furukawa K, Suzuki H, et al. Association of increased hepatic insulin clearance and change in serum triglycerides or β-hydroxybutyrate concentration via the sodium/glucose-cotransporter 2 inhibitor tofogliflozin. Diabetes, Obesity & Metabolism. 2020; 22: 947–956.
[122]
Bril F, Lomonaco R, Orsak B, Ortiz-Lopez C, Webb A, Tio F, et al. Relationship between disease severity, hyperinsulinemia, and impaired insulin clearance in patients with nonalcoholic steatohepatitis. Hepatology. 2014; 59: 2178–2187.
[123]
Ekholm E, Hansen L, Johnsson E, Iqbal N, Carlsson B, Chen H, et al. Combined treatment with saxagliptin plus dapagliflozin reduces insulin levels by increased insulin clearance and improves β-cell function. Endocrine Practice. 2017; 23: 258–265.
[124]
Newman JC, Verdin E. β-Hydroxybutyrate: A Signaling Metabolite. Annual Review of Nutrition. 2017; 37: 51–76.
[125]
Kanbay M, Bulbul MC, Copur S, Afsar B, Sag AA, Siriopol D, et al. Therapeutic implications of shared mechanisms in non-alcoholic fatty liver disease and chronic kidney disease. Journal of Nephrology. 2021; 34: 649–659.
[126]
Sarafidis P, Ferro CJ, Morales E, Ortiz A, Malyszko J, Hojs R, et al. SGLT-2 inhibitors and GLP-1 receptor agonists for nephroprotection and cardioprotection in patients with diabetes mellitus and chronic kidney disease. A consensus statement by the EURECA-m and the DIABESITY working groups of the ERA-EDTA. Nephrology, Dialysis, Transplantation. 2019; 34: 208–230.
[127]
Vallon V, Gerasimova M, Rose MA, Masuda T, Satriano J, Mayoux E, et al. SGLT2 inhibitor empagliflozin reduces renal growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. American Journal of Physiology. Renal Physiology. 2014; 306: F194–204.
[128]
Zhao Y, Xu L, Tian D, Xia P, Zheng H, Wang L, et al. Effects of sodium-glucose co-transporter 2 (SGLT2) inhibitors on serum uric acid level: A meta-analysis of randomized controlled trials. Diabetes, Obesity & Metabolism. 2018; 20: 458–462.
[129]
Scheen AJ, Esser N, Paquot N. Antidiabetic agents: Potential anti-inflammatory activity beyond glucose control. Diabetes & Metabolism. 2015; 41: 183–194.
[130]
Palmer SC, Tendal B, Mustafa RA, Vandvik PO, Li S, Hao Q, et al. Sodium-glucose cotransporter protein-2 (SGLT-2) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists for type 2 diabetes: systematic review and network meta-analysis of randomised controlled trials. British Medical Journal. 2021; 372: m4573.
[131]
Zhuo M, Hawley CE, Paik JM, Bessette LG, Wexler DJ, Kim DH, et al. Association of Sodium-Glucose Cotransporter-2 Inhibitors With Fracture Risk in Older Adults With Type 2 Diabetes. JAMA Network Open. 2021; 4: e2130762.
[132]
Dong B, Lv R, Wang J, Che L, Wang Z, Huai Z, et al. The Extraglycemic Effect of SGLT-2is on Mineral and Bone Metabolism and Bone Fracture. Frontiers in Endocrinology. 2022; 13: 918350.
[133]
Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. The New England Journal of Medicine. 2019; 380: 2295–2306.
[134]
Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, et al. Dapagliflozin in Patients with Chronic Kidney Disease. The New England Journal of Medicine. 2020; 383: 1436–1446.
[135]
Chertow GM, Vart P, Jongs N, Toto RD, Gorriz JL, Hou FF, et al. Effects of Dapagliflozin in Stage 4 Chronic Kidney Disease. Journal of the American Society of Nephrology. 2021; 32: 2352–2361.
[136]
Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. The New England Journal of Medicine. 2020; 383: 1413–1424.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top