Next Article in Journal
Organic Aciduria Disorders in Pregnancy: An Overview of Metabolic Considerations
Next Article in Special Issue
Metabolites Potentially Derived from Gut Microbiota Associated with Podocyte, Proximal Tubule, and Renal and Cerebrovascular Endothelial Damage in Early Diabetic Kidney Disease in T2DM Patients
Previous Article in Journal
Gymnema Sylvestre Supplementation Restores Normoglycemia, Corrects Dyslipidemia, and Transcriptionally Modulates Pancreatic and Hepatic Gene Expression in Alloxan-Induced Hyperglycemic Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potentials of Reducing Liver Fat in Non-Alcoholic Fatty Liver Disease: Close Association with Type 2 Diabetes

1
Division of Gastroenterology, Norfolk and Norwich University Hospital, Norwich NR4 7UY, UK
2
Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
3
Sports Medicine Laboratory, School of Physical Education & Sport Science, Aristotle University of Thessaloniki, 57001 Thessaloniki, Greece
4
Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Metabolites 2023, 13(4), 517; https://doi.org/10.3390/metabo13040517
Submission received: 4 March 2023 / Revised: 21 March 2023 / Accepted: 29 March 2023 / Published: 4 April 2023
(This article belongs to the Special Issue Metabolism in Diabetes Progression and Diabetic Complications)

Abstract

:
Nonalcoholic fatty liver disease (NAFLD), the most widespread chronic liver disease worldwide, confers a significant burden on health systems and leads to increased mortality and morbidity through several extrahepatic complications. NAFLD comprises a broad spectrum of liver-related disorders, including steatosis, cirrhosis, and hepatocellular carcinoma. It affects almost 30% of adults in the general population and up to 70% of people with type 2 diabetes (T2DM), sharing common pathogenetic pathways with the latter. In addition, NAFLD is closely related to obesity, which acts in synergy with other predisposing conditions, including alcohol consumption, provoking progressive and insidious liver damage. Among the most potent risk factors for accelerating the progression of NAFLD to fibrosis or cirrhosis, diabetes stands out. Despite the rapid rise in NAFLD rates, identifying the optimal treatment remains a challenge. Interestingly, NAFLD amelioration or remission appears to be associated with a lower risk of T2DM, indicating that liver-centric therapies could reduce the risk of developing T2DM and vice versa. Consequently, assessing NAFLD requires a multidisciplinary approach to identify and manage this multisystemic clinical entity early. With the continuously emerging new evidence, innovative therapeutic strategies are being developed for the treatment of NAFLD, prioritizing a combination of lifestyle changes and glucose-lowering medications. Based on recent evidence, this review scrutinizes all practical and sustainable interventions to achieve a resolution of NAFLD through a multimodal approach.

1. Putting into Context the Silent Epidemic: The Rise of NAFLD

Before the middle of the last decade, the worldwide prevalence of non-alcohol fatty liver disease (NAFLD) was approximately 25% in adult individuals. NAFLD is currently defined as an ectopic accumulation of lipids in the liver in the absence of secondary causes or other etiologies of liver disease [1,2]. It is histologically classified into two types: nonalcoholic fatty liver, defined as 5% liver steatosis with no evidence of injury to hepatocytes and no evidence of fibrosis; and nonalcoholic steatohepatitis (NASH), characterized as 5% liver steatosis, including inflammation and injury to hepatocytes with or without fibrosis [3]. NASH is estimated to be present in up to 60% of patients with NAFLD confirmed by biopsy [1]. Interestingly, while NAFLD was formally described more than 40 years ago, it has only been recognized in recent years as an important risk factor for metabolic disorders, related to the increasing burden of non-communicable diseases (NCDs), such as type 2 diabetes mellitus (T2DM), obesity, cardiovascular disease, and malignancy [4,5]. All of the above independent risk factors for mortality and morbidity are associated with a high economic cost to health care systems.
In the 21st century, among other metabolic diseases, diabetes mellitus has become the main concern for societies and health care systems. In particular, T2DM is mentioned to affect 1 in 11 adults and up to 463 million people worldwide [6]. According to the most recent data from the International Diabetes Federation (IDF), 700 million people between the ages of 20 and 79 will live with T2DM in 2045, around the world [7]. This type of diabetes, previously known as adult-onset diabetes, is defined by the following criteria: random blood sugar test ≥ 200 mg/dL (or ≥11.1 mmol/L) in symptomatic individuals, fasting blood sugar test ≥ 126 mg/dL (or ≥7 mmol/L) in two separate tests, oral glucose tolerance test (OGTT) ≥ 200 mg/dL (or ≥11.1 mmol/L) after two hours, and glycated hemoglobin (HbA1c) ≥ 6.5% in two separate assessments [8]. Growing evidence suggests that there is a strong positive association between NAFLD and diabetes mellitus, and, more specifically, individuals with diagnosed NAFLD have a twofold increased risk of T2DM [9]. This can be explained by the fact that the risk factors for diabetes overlap with the factors that affect the accumulation of liver fat. Thus, robust evidence from several epidemiological studies has reported that the detection of NAFLD in its early stages could predict the subsequent development of incident diabetes mellitus [10,11].

1.1. Pathophysiology of NAFLD/NASH

The development of NAFLD is an intricate process and is not completely understood. The liver, as is widely known, promotes many functions through the mobilization, regulation, and storage of nutrients. Hepatocytes play an essential role as regulators of amino acid, triglyceride and lipoprotein metabolism, gluconeogenesis, and ketogenesis [12]. Interestingly, although the liver is not the primary organ for lipid storage, there can be a few conditions that can cause ectopic lipid accumulation in it. The main causes include excessive intake of dietary fat, de novo lipogenesis, and hepatic uptake of non-esterified fatty acids from serum [12,13]. Insulin resistance causes lipolysis of adipose tissue, leading to a higher concentration of non-esterified fatty acids in the circulation, which are taken up by the liver in a concentration-dependent manner [14]. In addition, other metabolic disturbances, such as hyperinsulinemia and hyperglycemia, promote increased liver conversion of carbohydrates into fatty acids through de novo lipogenesis [15]. It is worth noting that the expression and action of various enzymes involved in lipogenesis and fatty acid storage in triglycerides are increased in individuals with NAFLD [16]. These enzymes, including diacylglycerol acyltransferase, stearoyl-CoA desaturase, fatty acid synthase, ketohexokinase, and acetyl-CoA carboxylase, have been explored as potential therapeutic weapons against NAFLD.
In a small percentage of patients, NAFLD is associated with infectious pathologies that can lead to the appearance of liver steatosis, such as hepatitis C and the human immunodeficiency virus (HIV). In some cases, it is related to medications (e.g., glucocorticoids, tamoxifen, tetracycline, total parenteral nutrition, amiodarone, methotrexate, valproic acid, and vinyl chloride) and specific toxins or acquired/inherited metabolic diseases such as cachexia, lipodystrophy, or even gastrointestinal surgery [17,18].
The development of NASH has been reported to be divided into two phases. The first is fatty deposition in the liver combined with an increase in insulin resistance. The second phase is related to cellular and molecular changes, including primarily oxidative stress and oxidation of fatty acids through lipid peroxidation, hyperinsulinemia, energy homeostasis, variation in the extracellular matrix, and changes in immune system function [19,20]. Consequently, the development of insulin resistance is even more complex than it appears. Although increased fat mass and adipocyte differentiation play a key role in the development of insulin resistance, the relationship between glycemia and the resolution of existing NAFLD/NASH (defined by the absence of ultrasound criteria for NAFLD/NASH on repeated imaging) is still a knowledge gap.

1.2. Unravelling the Connection: Pathophysiological Links between NAFLD and Diabetes Mellitus

Robust evidence suggests that there is a strong and bidirectional relationship between the progression of NAFLD and T2DM. High levels of diacylglycerol or ceramides can affect liver–insulin signaling, leading to an abnormal increase in liver insulin resistance [21]. Furthermore, elevated concentrations of circulating transaminases are strongly associated with a future higher risk of T2DM [22]. Except for the above, patients with NAFLD and T2DM are at an increased risk of developing macrovascular and microvascular diseases such as cardiovascular and chronic kidney disease, which are the main causes of mortality in these individuals [23]. Due to the intricate inter-relationship and the high global prevalence of NAFLD and T2DM, targeting insulin sensitivity and hyperglycemia combined with weight loss and adopting a holistic approach to the treatment of metabolic disease in patients with NAFLD could prove advantageous. Therefore, it has been suggested that alleviating NAFLD and NASH should be considered as part of the therapeutic strategy in patients with T2DM [24].
It should be noted that several studies have shown that improvements in insulin sensitivity have been positively associated with histological improvements in NAFLD/NASH and regression of fibrosis [25]. In 2013, Sung et al. reported that the risk of T2DM decreased in patients with resolved NAFLD status, as NAFLD is a reversible condition in its early stages [26]. A meta-analysis, published in 2018, found that the severity of NAFLD is directly related to dysglycemia, having a significant impact on the future risk of developing T2DM [23]. However, more studies are needed to prove the causal relationship between the two conditions and to reveal the extent of the risk of T2DM caused by the variable stages of NAFLD.
The purpose of this narrative review is to summarize the current literature on modern therapeutic approaches to NAFLD in patients with and without diabetes, focusing on the targeting of metabolic disturbances. Another purpose is to discuss future potential treatments and knowledge gaps in the therapy of NAFLD.

2. Pathways to Healing: Navigating the Therapeutic Approach

The therapeutic quiver of NAFLD consists of several levels, of which lifestyle, pharmaceutical, and surgical approaches are the main treatments. A multimodal intervention with multiple aspects, such as lifestyle modification, weight loss, specific diets, and medication, is the most appropriate and holistic approach for most people with NAFLD.

2.1. Less Is More: Unlocking the Power of Conservative Treatment

2.1.1. The Power of Lifestyle Modification in NAFLD

Robust evidence supports the crucial role of lifestyle changes as primary options for the treatment of NAFLD. These approaches, which include diet, exercise, or physical activity, mainly aim to control metabolic status [27]. Looking back in 2004, the World Health Organization (WHO) established that moderate intensity exercise improves not only physical and mental health, but also metabolic syndrome, T2DM, and cardiovascular disease, conditions that are inseparably related to NAFLD [28]. In the 21st century, physical activity is considered a pillar determinant of metabolic control and is recommended for NAFLD. Different types of exercise, such as high-intensity intermittent exercise, aerobic exercise, or resistance exercise, seem to have beneficial effects on fatty liver disease [29,30]. In 2017, Oh et al. found that high-intensity interval aerobic exercise, moderate-intensity continuous aerobic exercise, and resistance exercise were equally effective in decreasing liver fat content. However, only high-intensity interval aerobic exercise had a beneficial effect on restoring Kupffer cell function [31]. A randomized control trial by Zhang et al. indicated that after a 12-month active intervention, the two exercise groups (strong and moderate training) showed a significant reduction in intrahepatic triglyceride content (measured by proton magnetic resonance spectroscopy) compared to the control group [32]. Furthermore, several studies reported that resistance training leads to a reduction in liver fat of 4–47% independently of weight loss [33]. The mechanisms underpinning the reduction of hepatic fat deposition due to exercise reflect changes in insulin sensitivity and circulatory lipids. Exercise not only improves glycemic control but suppresses de novo lipogenesis and improves blood pressure levels in people with NAFLD [34,35,36]. Table 1 summarizes key studies on the effect of different exercises in patients with NAFLD.

2.1.2. Shedding Pounds: The Science of Weight Loss and Calorie Restriction

Weight loss is a gold standard therapy for most patients with NAFLD and can regress liver disease, along with the reduction of cardiovascular disease and the risk of T2DM [46]. Some researchers support that a weight reduction of 10% is capable of inducing the resolution of NASH and improving fibrosis by at least one stage [47]. According to the guidelines of the American Association for the Study of Liver Diseases (AASLD), a weight loss of 5–10% in overweight or obese individuals and 3–10% in non-obese individuals with NAFLD is the primary objective of lifestyle interventions. In accordance with the above, there are also the National Institute of Health and Care Excellence (NICE) guidelines [48,49]. In addition, obesity, as a result of excess caloric consumption, is one of the leading factors for NAFLD. Caloric restriction acts on metabolic reprogramming and on the utilization of body energy, reducing oxidative damage to cells [50]. Because carbohydrates (which are the main energy source of the human body) are linked to NAFLD, their restriction in the diet can lead to lower glycemic load, increased insulin sensitivity, and pancreatic β-cell insulin secretion of pancreatic cells [51].
A clinical trial by Holmer et al., which recruited 74 patients with NAFLD, indicated that intermittent calorie restriction and a low carbohydrate high fat diet (LCHF) are more effective in reducing liver steatosis and body weight compared to general lifestyle modification. Participants were randomized into 3 groups: intermittent calorie restriction, including 500 kcal/day for women and 600 kcal/day for men; LCHF, with an average daily calorie intake of 1600 kcal/day for women and 1900 kcal/day for men; and general lifestyle advice [52]. Furthermore, in a prospective study by Vilar et al., a combination of exercise and a hypocaloric diet revealed a dose–response relationship between weight reduction and general histological parameters, with the greatest improvement detected in those with the greatest weight loss [53]. However, the beneficial effects of a low-carbohydrate diet are only in the short term. In the long term, a reduced carbohydrate and a reduced fat diet has results similar to those who achieved a 7% weight loss [54]. Table 2 presents the effect of dietary intervention on NAFLD.
In addition to the above, it should be noted that some studies reported the beneficial effect of diabetes remission on NAFLD and pancreatic morphology. In 2020, a post hoc analysis of the DiRECT trial showed changes in the gross morphology of the pancreas 2 years post T2DM remission. The size of the pancreas had increased in patients who achieved remission and weight loss, compared to those who did not respond to the weight loss intervention. Intrahepatic fat and levels of FGF-21 and FGF-19 also decreased. However, it is notable that there is no significant increase in pancreas volume after 6 months of reversal of type 2 diabetes [55]. Additional trials might be of interest from a scientific point of view to investigate further data on the progression of NAFLD and changes in pancreatic tissue after remission of diabetes.

2.1.3. Breaking the Link between Fatty Liver and Type 2 Diabetes: The Power of Nutritional Interventions

Numerous studies have corroborated the pivotal role of certain macronutrients in the initiation and progression of NAFLD, regardless of caloric intake. In particular, macronutrients such as saturated fatty acids (SFA), trans fats, simple sugars such as sucrose and fructose, and animal proteins are known to inflict damage on the liver through the accumulation of triglycerides and impaired antioxidant activity, compromising insulin sensitivity and postprandial triglyceride metabolism [56]. In contrast, the consumption of monounsaturated fatty acids (MUFA), ω3 polyunsaturated fatty acids (PUFA), plant-based proteins, and dietary fibers such as whole grain cereals, fruits and vegetables, fatty fish (which are primarily rich in ω3), and extra virgin olive oil have been found to confer beneficial effects [57,58]. Gupta et al. suggest that oily fish (2–4 g/d), coffee (≥3 cups/day), and nuts (100 g/d) are recommended as suitable additions to physical activity and caloric restriction for patients with fatty liver disease, based on strong evidence from human trials. Although tea, red wine, avocado and olive oil can be consumed moderately without harm, more research is needed to investigate their therapeutic benefits for patients with NAFLD/NASH [59].
Furthermore, Halima et al. conducted a study investigating the impact of apple cider vinegar on rats with diabetes and demonstrated that in addition to its potent antihyperglycemic properties, it also exhibited a crucial hepatoprotective effect. In particular, indicators of liver toxicity, namely ALT, AST, total and direct bilirubin, as well as levels of TC, TG, and LDL-c, demonstrated a significant reduction, which was particularly prominent after four weeks of treatment, together with an elevation in HDL-c [60]. These findings are consistent with several other studies [61,62,63]. The above elucidated results unequivocally demonstrate that daily ingestion of vinegar can mitigate the increase in blood glucose levels and lipid profile, which is typically induced by a hypercaloric diet in rats, as posited by Ousaaid et al. [64]. Therefore, the use of apple cider vinegar could confer considerable advantages in avoiding metabolic irregularities commonly associated with a high-calorie diet.
Table 2. Dietary interventions and outcomes in NAFLD patients.
Table 2. Dietary interventions and outcomes in NAFLD patients.
StudyType of StudyNumber of PatientsType of DietPatients with NAFLDDurationInsulin ResistanceOutcomes
Haufe et al. (2011) [65]RCT52 vs. 50Hypocaloric LCD vs. LFDOverweight/obese6 months↓ to a
similar extent
42% vs. 47% ↓ in IHLC
Browning et al. (2011) [66]Non-RCT9 vs. 9VLCD vs. Hypocaloric dietWithout cirrhosis2 weeksNot evaluated55% vs. 28% ↓ in IHLC
Ryan et al. (2013) [67]RCT6 vs. 6Med. vs. LFD or HCDBiopsy-proven6 weeks↓ with the
Med. diet
39% vs. 7% ↓ of IHLC
Vilar-Gomez et al. (2015) [53]Single-arm261Hypocaloric LFD + PAHistological NASH52 weeksAssociations between
weight loss and
histological improvement
Misciagna et al. (2017) [68]RCT44 vs. 46Med. vs. CDModerate-severe (US)6 monthsImprovementSignificant improvement of NAFLD score
Abenavoli et al. (2017) [69]RCT20 vs. 20 vs. 10Med. ± Antioxidant supplementation (1400–1600 kcal/d) vs. CDOverweight6 months↓ of insulin resistance and fasting glucose↓ of FLI and LSM in both diets
Markova et al. (2017) [70]RCT18 vs. 19Isocaloric animal-protein
vs. plant-protein diet
T2DM6 weeks↓ of insulin resistance and fasting glucose48% vs. 35.7% ↓ in IHLC
Katsagoni et al. (2018) [71]RCT21 vs. 21 vs. 21Hypocaloric Med. vs. Med. + lifestyle intervention vs. CDOverweight/obese6 monthsNot evaluated↓ of LSM in both diets, improvement in ALT only in Med. + lifestyle intervention-group
Marin-Alejandre et al. (2019) [72]RCT37 vs. 39Hypocaloric diet vs. CDOverweight/obese6 monthsSignificant reduction in glucose and insulin↓ in IHLC + FLI following both diets
Gepner et al. (2019) [73]RCT76 vs. 63 vs. 73
vs. 66
LFD vs. LFD with PA vs. Med./LCD vs. Med./LCD with PAAbdominal obesity18 monthsSignificant reduction in glucose and insulin7.3% (Med./LCD) vs. 5.8 (LFD) ↓ in IHLC after 6 months,
4.2% vs. 3.8% after 18 months
Yaskolka Meir et al. (2020) [74]RCT89 vs. 84 vs. 91Hypocaloric Med. (1500–1800 kcal/d ♂, 1200–1400 kcal/d ♀ vs. healthy dietAbdominal obesity18 monthsNot evaluated↓ IHLC following all diets
Xu et al. (2020) [75]RCT10 vs. 9 vs. 10Hypocaloric LPD vs. HPD
vs. reference-protein
diet
Obese3 weeksNot evaluated36.7% vs. 42.6% ↓ in IHLC vs. no changes in IHLC
Goss et al. (2020) [76]RCT14 vs. 11LCD vs. LFDObese8 weeks↓ of insulin resistanceNo significant
difference
Holmer et al. (2021) [52]RCT20 vs. 24 vs. 20LCD vs. 5:2 diet vs. CDNAFLD12 weeks↓ of insulin resistance and HbA1c53.1% vs. 50.9% vs. 16.8% ↓ in
IHLC, 61.9% vs. 63.8% vs. 20.2% ↓ in CAP, change in IHLC 3.9%
greater in LCD compared to CD and 2.6% in 5:2 diet compared to CD, ↓ in LSM in 5:2 diet and CD compared to
LCD
CAP: controlled attenuation parameter, CD: control diet, NAFLD: non-alcoholic fatty liver disease, NASH: non-alcoholic steatohepatitis, LCD: low-carbohydrate diet, LFD: low-fat diet, PA: physical activity, HPD: high-protein diet, LPD: low-protein diet, LSM: liver stiffness measurement, IHLC: intrahepatic lipid content, LFD: low-fat diet, Med.: Mediterranean diet, FLI: fatty liver index, ↓: decrease, ♂: male, ♀: female

2.2. Cutting-Edge Solutions: Exploring Surgical Therapies for NAFLD

The potential effects of bariatric surgery on liver fat disease may extend beyond weight loss. In fact, serum concentrations of glucagon-like peptide-1 (GLP-1) increase after metabolic surgery, leading to decreased appetite, slower gastric emptying, and improved insulin sensitivity [77]. Furthermore, the main role of GLP-1 is to modulate bile acid signaling through the farnesoid X receptor (FXR), which can modify the gut microbiome and promote NAFLD [78]. Therefore, current guidelines recommend that metabolic surgery can be a potential approach in patients with T2DM or overweight/obese individuals (i.e., BMI > 35 kg/m2) [48,79]. Although bariatric surgery has a beneficial effect, various limitations such as patient acceptability of complications, availability of services, and high cost make its use difficult and highlight the need to carefully select eligible candidates [80].
The most common bariatric surgery procedures include adjustable gastric band (AGB), biliopancreatic diversion (BPD), vertical sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB). Consequently, different methods might induce variable biological effects depending on the surgical procedure. The most common metabolic operations are SG and RYGB. In the first, about 80% of the stomach portion is removed along the gastric greater curvature and the small dimensions of the stomach, along with the changes in the hormonal environment, reduce hunger and delay gastric emptiness. In the second procedure, the stomach is separated into a smaller pouch in the smaller curvature (through stapling) and anastomosed with the jejunum [81,82]. In fact, by restricting food intake and by promoting malabsorption of nutrients, these techniques can cause weight loss. Both the reduction in body weight and decrease in waist circumstance through bariatric surgery led to improvement in insulin resistance, T2DM, obesity, fatty liver disease, and dyslipidemia [80]. Interestingly, one of the most important outcomes of bariatric surgery is that it can markedly improve all histological characteristics of NAFLD, including fibrosis. According to Lee et al., a resolution of steatosis was observed in 66% of patients, a resolution of inflammation in 50% of patients, and a resolution of fibrosis in 40% of patients after bariatric surgery [83]. Another recent study published in 2018, showed a resolution of NAFLD of 78% after RYGB [84]. Furthermore, Weiner reported that patients after AGB, RYGB and BPD achieved complete regression of NAFLD in up to 82.8% of the cases [85].
Table 3 presents studies that have investigated the effectiveness of bariatric surgery management in alleviating NAFLD.

2.3. Exploring the Innovative World of Pharmaceutical Solutions

2.3.1. Effects of Anti-Diabetic Agents on NAFLD

It is well established that fatty liver and T2DM are the two sides of the same coin, sharing common pathogenic pathways and factors such as insulin resistance. Although the coexistence of fatty liver and T2DM is increasing, the treatment is not adequate. Due to this close link between T2DM and NAFLD, various glucose-lowering drugs have been used as NAFLD therapeutics. Numerous clinical trials have shown the beneficial effects of GLP-1 receptor agonists, insulin-sensitizing thiazolidinediones, and sodium-glucose cotransporter 2 (SGLT2) inhibitors on liver fat content. GLP-1 binds to a specific GLP-1 receptor, whose activation can promote the reduction of liver steatosis by improving insulin signaling pathways, hepatocyte lipotoxicity, and mitochondrial function [95,96]. On the contrary, SGLT2 inhibition promotes negative energy balance through increased glycosuria and a change of the substrate to lipids as an energy source, which inhibit liver steatosis, inflammation, and fibrosis [97]. In 2021, a meta-analysis by Mantovani et al. showed that treatment with GLP-1 receptor agonists or SGLT2 inhibitors compared to placebo decreased the absolute percentage of liver fat content and serum levels of ALT [98]. These findings have been replicated by several other studies [99].
It is significant to mention that a multitude of studies have recently elucidated the efficacy of SGLT2 inhibitors in alleviating liver steatosis. Of particular interest, in 2018, Shibuya et al. revealed, for the first time, the statistically significant and advantageous impact of luseogliflozin, compared to metformin, on NAFLD and weight loss [100]. The results manifested a superior effect of the former, particularly on body mass index (BMI), after a six-month period. Luseogliflozin facilitates the mitigation of visceral adiposity by excreting energy through urine glucose excretion [100]. Additionally, in the same year, a double-blind randomized controlled study revealed that dapagliflozin monotherapy can reduce the levels of hepatocyte injury biomarkers, such as ALT, AST, γ-glutamyl transferase (γ-GT), cytokeratin 18-M30, cytokeratin 18-M65, and plasma fibroblast growth factor 21 (FGF21). With the combination of omega-3 carboxylic acids, not only can glucose control be improved, but body weight and abdominal fat volumes can also be reduced [101]. Analogous results were observed in a study by Sattar et al. using empagliflozin, in which the decline in ALT was consistently more notable than that of AST. The reductions were most prominent in participants with the highest baseline ALT levels and were primarily unaffected by changes in HbA1c and body weight [102]. However, the exact mechanisms through which empagliflozin mitigates aminotransferases or liver fat remain ambiguous; therefore, more research is needed.
Thiazolidinediones increase peripheral insulin sensitivity by stimulating adipokines, promoting triglyceride storage in adipose tissue, and improving the suppressive action of insulin on lipolysis [103]. In this way, thiazolidinediones lead to lower serum levels of free fatty acids and reduced hepatic lipid accretion [103]. The most common and approved are pioglitazone and rosiglitazone, which are powerful activators of the nuclear receptor PPARγ and are expressed mainly in adipose tissue [104]. Numerous studies have shown that pioglitazone has a beneficial effect on insulin sensitivity, inflammation, and hepatocyte degeneration, but there was no difference in the resolution of fibrosis [105]. A recent meta-analysis of randomized controlled trials using pioglitazone or rosiglitazone revealed that both drugs led to enhanced liver histology, including decreased steatosis, inflammation, and hepatocyte degeneration [106].
Recently, semaglutide and tirzepatide have been added to the therapeutic quiver against T2DM and obesity. Semaglutide is a GLP-1 analogue, and tirzepatide is a dual analogue of GLP-1 and GIP (glucose-dependent insulinotropic polypeptide) [107]. Both delivered impressive results in the phase 3 trials and can be considered future game changers in the realm of T2DM remission. Due to their importance in NASH therapy, these agents will be discussed in detail at a later part of the review. Table 4 summarizes the clinical trials that have examined the effects of antidiabetic drugs on NAFLD.

2.3.2. Effects of Statins and Other Lipid-Lowering Drugs

Realizing that NALFD is strongly related to metabolic syndrome, there is a need for an integrated approach for individuals with a high liver fat content. The definition of metabolic syndrome includes the following criteria: abdominal obesity, with waist circumference of ≥90 cm in men or ≥85 cm in women; low high-density lipoprotein (HDL) cholesterol with HDL-cholesterol of <40 mg/dL in men and <50 mg/dL in women; hypertriglyceridemia with triglyceride (TG) of ≥150 mg/dL; high systolic blood pressure (BP), with systolic BP of ≥130 mmHg and/or diastolic BP of ≥85 mmHg; or hyperglycemia, with fasting plasma glucose (FPG) of >100 mg/dL [125,126]. Consequently, abnormal blood cholesterol levels play a key role in the progression of NAFLD and can be controlled with statins [127]. Except for this action, statins exhibit pleiotropic properties, such as antioxidant and anti-inflammatory effects, neoangiogenesis, and improvement of endothelial functions [127]. Interestingly, a large amount of data recommended that a statin remedy is correlated with a significant improvement in liver steatosis, inflammation, and even fibrosis [128,129]. For example, an observational study by Lee et al. found a lower risk of NAFLD in patients who received statin therapy [129]. According to a meta-analysis of six studies, ezetimibe, which is a lipid lowering agent acting by reducing cholesterol absorption in the intestines, significantly reduced plasma liver enzyme levels, as well as improved liver steatosis [130]. On the contrary, fenofibrate, a PPAR-a agonist, does not have a significant effect on liver fat content [131].
Furthermore, the category of omega-3 polyunsaturated fatty acids (n-3 PUFAs), such as a-linolenic acid (a-ALA), stearidonic acid (SDA), eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA), has a beneficial effect on peripheral insulin sensitivity and on triglycerides levels, leading to a lower deposition of liver fat [132]. A randomized control trial published in 2020 reported lower liver fat in patients who received n-3 PUFA supplementation compared to those who received the placebo [132]. Similar outcomes were observed in another meta-analysis [133]. However, more well-designed randomized clinical trials are necessary to suggest omega-3 PUFA supplementation for the treatment of NAFLD in patients with and without T2DM.
Although statins have long been used as a widely accepted method for reducing cholesterol and minimizing the risk and mortality associated with cardiovascular disease, recent years have seen increasing scrutiny of their potential side effects. It should be noted, in particular, the increasing evidence linking prolonged statin use with diabetes progression and ectopic fat deposition, particularly in the kidneys of patients with diabetic nephropathy [134]. Recently, a study by Huang et al. revealed that statin administration for a period of more than 10 years was found to increase insulin resistance, alter lipid metabolism, provoke inflammation and fibrosis, and ultimately exacerbate the progression of diabetic nephropathy in diabetic mice. It is worth noting that the duration of the study spanned 50 weeks, which is equivalent to at least 35 years in the human life cycle [134]. Similar results were reported in the retrospective cohort study by Mansi et al., which followed patients with diabetes for a 12-year period and highlighted the need to carefully consider the metabolic effects of statin use when evaluating its risk–benefit ratio for diabetic individuals [135]. However, these findings are in stark contrast to the beneficial effects of statins in the treatment of NAFLD. Future research efforts are expected to provide more information on whether prolonged statin use may yield more detrimental than advantageous outcomes.

2.3.3. From Lab to Liver: The Promising Future of Developing New Drugs for NAFLD

More recently, new potential drugs for NAFLD have been studied. The most common treatment targets are the farnesoid X receptor (FXR), a nuclear receptor, and obeticholic acid (OCA), which is a synthetically modified analogue of chenodeoxycholic acid [136]. These agents improve insulin resistance, regulate glucose and lipid metabolism, and have anti-inflammatory and anti-fibrotic effects in NAFLD [137]. Yonoussi et al. observed that 308 patients who received OCA 25 mg daily had an improvement in fibrosis, compared to the control group [138]. Interestingly, the multicenter, randomized, placebo-controlled FLINT trial documented that those who responded to OCA, defined as patients with a ≥30% reduction in liver lipid content, had an improvement in the histological features of NASH, including fibrosis and steatosis as well as reduced cell death [139]. However, one of the limitations of OCA use is that it can increase serum LDL levels. Therefore, the safety of the combination of OCA with statin in patients with NASH is being tested in ongoing studies. Non bile acid farnesoid X-activated receptor (FXR) agonists, including tropifexor, cilofexor, EDP-305, and nidufexo, have also been tested for NAFLD [140]. The main difference between OCA and cilofexor is that the latter caused a reduction in liver lipid content in patients with NASH without altering blood levels of lipids or indicators of insulin resistance [141]. An anti-inflammatory drug, the stearoyl-CoA desaturase (SCD) 1 inhibitor, plays a key role in liver lipogenesis. The main action is to catalyze the conversion of saturated fatty acids to MUFA, protecting against hepatic steatosis [142,143]. Two animal studies showed that SCD1 activity was elevated in proportion to liver lipid content in models of NAFLD and genetic knockout of hepatic SCD1 expression effectively reduced fatty liver and insulin resistance in animals fed a high-fat diet [144,145]. Recently, a clinical trial revealed a reduction in liver fat content, resolution of NASH, and improvement of liver fibrosis in individuals with NAFLD and prediabetes or T2DM after the use of aramchol [146]. Given that to date there is no FDA-approved therapy for the treatment of NAFLD, evaluating the efficacy and safety profile of new agents is of paramount importance. However, the fact that liver biochemistry does not always reflect hepatic histology makes the conduction of biopsy studies important, which on the other hand, present significant technical challenges for obvious reasons.
Table 5 presents ongoing trials investigating the safety and efficacy of new medications for the treatment of NAFLD.

2.3.4. Effects of Anti-Obesity Drugs

During recent years, weight loss pharmacotherapy has been an attractive option for patients with NAFLD, with or without T2DM and with a BMI > 30 kg/m2 or >27 kg/m2 in the presence of at least one metabolic comorbidity, because it can lead to the remission of diabetes and improves the progression of fatty liver disease [143,146]. Τhe US Food and Drug Administration (FDA) has approved six medications for chronic weight management: orlistat, lorcaserin, phentermine/topiramate, bupropion/noltrexone, liraglutide and semaglutide that are associated with a decrease in body weight of at least 5% in one year [146]. Interestingly, a closer look at the literature reveals that, except for the long-term effects on glycemic control, orlistat selectively reduces visceral fat and prevents the digestion of free fatty acids that are responsible for the increase in liver and peripheral insulin resistance [147]. In 2005, a meta-analysis of seven randomized control trials showed that patients who received 120 mg orlistat three times received an average weight loss of 3.8 kg compared to 1.4 kg in the placebo group after 12 weeks [148]. In contrast, no data are available on the effects of topiramate, naltrexone, bupropion, and phentermine on liver outcomes in patients with NAFLD [149].

3. Tirzepatide and Semaglutide: New Weapons against NAFLD

As previously stated, two recently approved drugs, tirzepatide, a dual analogue of GIP/GLP-1, and semaglutide, a GLP-1 analogue, were added to the therapeutic arsenal of NAFLD [150]. A phase 3 trial reported that the liver fat content was significantly reduced after tirzepatide therapy compared to baseline measures [151]. On the other hand, a phase 2 trial involving patients with NASH revealed that semaglutide treatment resulted in a significantly higher percentage of patients with NASH resolution than the placebo [113].
Tirzepatide is associated with a significant reduction in liver fat content (LFC), subcutaneous abdominal adipose tissue (ASAT), and volume of visceral adipose tissue (VAT) [152,153]. A multicenter phase 3 clinical trial reported a significant reduction in LFC, ASAT, and VAT volumes compared to insulin degludec in individuals with T2DM [151]. The results were more prominent in groups receiving 10 mg and 15 mg of tirzepatide. An MRI scan was performed in participants before randomization and the primary efficacy endpoint was identified by MRI-proton density fat fraction (MRI-PDFF) at week 52 of drug administration [151]. According to a meta-analysis of 11 phase 2 randomized controlled trials, the treatment regimen with a GLP-1 receptor agonist (liraglutide or semaglutide) for a median of 26 weeks was strongly associated with significant reductions in LFC, as well as increased histological resolution of NASH without worsening of fibrosis, compared to the placebo or reference therapy group [154].
In 2020, Hartman et al. determined the result of tirzepatide on the biomarkers of NASH and fibrosis in people with diabetes. A reduction in ALT, AST, keratin-18 (K-18), and N-terminal pro-peptide of type III collagen (Pro-C3) levels was revealed, combined with a significant increase in adiponectin levels that was dose-dependent [155]. It is a well-established fact that K-18 and Pro-C3 serve as biomarkers of fibrogenesis in individuals afflicted with NAFLD. As mentioned above, to date, there has not been approved pharmacological therapy to resolve NAFLD. However, the remarkable effects of tirzepatide on LFC promise that it can be more effective than GLP-1 receptor agonists, but the preliminary promising findings should be confirmed by further studies. An ongoing trial, SYNERGY-NASH, will provide specific data on the beneficial effect of tirzepatide in participants with biopsy-proven NASH.
Semaglutide has also been effective in improving liver biochemistry. A prospective study by Volpe et al. showed a significant decrease in glucometabolic parameters, liver enzymes, and laboratory indicators of liver steatosis during therapy, as well as a reduction in fat mass and VAT. Interestingly, liver steatosis improved in 70% of participants after 52 weeks of treatment [156]. Furthermore, in 2021, a randomized control trial compared changes in liver stiffness and liver fat in individuals with NAFLD who received semaglutide and placebo therapy, respectively. The results revealed a significant reduction in liver steatosis with an improvement in liver enzymes and metabolic parameters in the semaglutide group versus the placebo group. However, no significant differences in liver stiffness were observed in both groups [157]. This year, Mantovani et al. published a systematic review providing data on the beneficial effect of GLP-1 receptor antagonists on the histological features of NAFLD, including steatosis, ballooning, and lobular inflammation, as well as on the resolution of NASH without worsening of fibrosis [158].
Recently, an open-label phase 2 trial by Alkhouri et al. reported that the combination of semaglutide with cilofexor and firsocostat achieved greater improvements in liver steatosis and biochemistry profile than monotherapy [159]. Based on the evidence mentioned above, semaglutide in combination with weight loss and a specific diet should be recommended as an optimal approach for the treatment of individuals with T2DM and fatty liver disease.
Future studies are expected to add additional evidence on the effects of new antidiabetic agents on NAFLD progression and clarify whether the benefits are driven primarily by weight loss or by the pleiotropic actions of the drugs.

4. Conclusions

The review of available evidence supports the notion that NAFLD is an important risk factor for the development of T2DM, but also underlines the bidirectional relationship between these conditions. Despite many advances in our knowledge on the epidemiology and pathogenesis of NAFLD, the most established treatment so far is intensive weight loss. The importance of weight reduction is highlighted in people with NASH, where weight loss greater than 7% is associated with a clinically significant regression of disease status [45]. Exercise compared to weight loss produces significant but modest changes in liver fat. Lifestyle modification, including specific diets and physical activity, is and should be the first line of treatment in NAFLD and NASH because it has been shown to ameliorate the risk of extrahepatic comorbidities and complications. On this basis, the current European and American clinical guidelines for the treatment of NAFLD have strongly suggested the importance of routine screening for T2DM in all patients with NAFLD [160].
Today, the question is no longer whether lifestyle modification is an effective clinical therapy, but how we implement lifestyle as a therapy in daily clinical care. However, most studies indicate several barriers to maintaining a healthy lifestyle in the long term and that weight regain is very common among people with diabetes and/or obesity, making the use of pharmacotherapy a necessity in most cases. In this context, a better understanding of the mechanisms linking NAFLD and T2DM will help design targeted therapies that can stop or reverse disease progression.

Author Contributions

G.T. (Georgios Tsamos), T.K. and D.V. reviewed the literature and drafted the first version of the manuscript. V.M., K.M. and G.T. (Georgios Tzimagiorgis) reviewed the literature and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

Conflicts of Interest

T.K. has received honoraria for lectures from AstraZeneca, Boehringer Ingelheim, Pharmaserve Lilly and Novo Nordisk, for advisory boards from Novo Nordisk and Boehringer Ingelheim, and has participated in sponsored studies by Eli-Lilly and Novo Nordisk. The other authors report no conflict of interest.

References

  1. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Li, J.; Zou, B.; Yeo, Y.H.; Feng, Y.; Xie, X.; Lee, D.H.; Fujii, H.; Wu, Y.; Kam, L.Y.; Ji, F.; et al. Prevalence, incidence, and outcome of non-alcoholic fatty liver disease in Asia, 1999–2019: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2019, 4, 389–398. [Google Scholar] [CrossRef] [PubMed]
  3. Takahashi, Y.; Fukusato, T. Histopathology of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J. Gastroenterol. 2014, 20, 15539–15548. [Google Scholar] [CrossRef] [PubMed]
  4. GBD 2016 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017, 390, 1211–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gakidou, E.; Afshin, A.; Abajobir, A.A.; Abate, K.H.; Abbafati, C.; Abbas, K.M.; Abd-Allah, F.; Abdulle, A.M.; Abera, S.F.; Aboyans, V.; et al. Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017, 390, 1345–1422. [Google Scholar] [CrossRef] [Green Version]
  6. International Diabetes Federation. IDF Diabetes Atlas Tenth Edition. 2021. Available online: https://diabetesatlas.org/ (accessed on 12 September 2021).
  7. 9th Edition|IDF Diabetes Atlas. Available online: https://diabetesatlas.org/atlas/ninth-edition/ (accessed on 1 March 2020).
  8. Kerner, W.; Brückel, J.; German Diabetes Association. Definition, Classification and Diagnosis of Diabetes Mellitus. Exp. Clin. Endocrinol. Diabetes 2014, 122, 384–386. [Google Scholar] [CrossRef] [Green Version]
  9. Ballestri, S.; Zona, S.; Targher, G.; Romagnoli, D.; Baldelli, E.; Nascimbeni, F.; Roverato, A.; Guaraldi, G.; Lonardo, A. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J. Gastroenterol. Hepatol. 2016, 31, 936–944. [Google Scholar] [CrossRef]
  10. Park, S.K.; Seo, M.H.; Shin, H.C.; Ryoo, J.-H. Clinical availability of nonalcoholic fatty liver disease as an early predictor of type 2 diabetes mellitus in korean men: 5-year prospective cohort study. Hepatology 2012, 57, 1378–1383. [Google Scholar] [CrossRef]
  11. Kim, S.S.; Cho, H.J.; Kim, H.J.; Kang, D.R.; Berry, J.R.; Kim, J.H.; Yang, M.J.; Lim, S.G.; Kim, S.; Cheong, J.Y.; et al. Nonalcoholic fatty liver disease as a sentinel marker for the development of diabetes mellitus in non-obese subjects. Dig. Liver Dis. 2018, 50, 370–377. [Google Scholar] [CrossRef]
  12. Donnelly, K.L.; Smith, C.I.; Schwarzenberg, S.J.; Jessurun, J.; Boldt, M.D.; Parks, E.J. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Investig. 2005, 115, 1343–1351. [Google Scholar] [CrossRef] [Green Version]
  13. Fabbrini, E.; Mohammed, B.S.; Magkos, F.; Korenblat, K.M.; Patterson, B.W.; Klein, S. Alterations in Adipose Tissue and Hepatic Lipid Kinetics in Obese Men and Women With Nonalcoholic Fatty Liver Disease. Gastroenterology 2008, 134, 424–431. [Google Scholar] [CrossRef] [Green Version]
  14. Korenblat, K.M.; Fabbrini, E.; Mohammed, B.S.; Klein, S. Liver, Muscle, and Adipose Tissue Insulin Action Is Directly Related to Intrahepatic Triglyceride Content in Obese Subjects. Gastroenterology 2008, 134, 1369–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Smith, G.; Shankaran, M.; Yoshino, M.; Schweitzer, G.G.; Chondronikola, M.; Beals, J.W.; Okunade, A.L.; Patterson, B.W.; Nyangau, E.; Field, T.; et al. Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease. J. Clin. Investig. 2020, 130, 1453–1460. [Google Scholar] [CrossRef]
  16. Samuel, V.T.; Shulman, G.I. Nonalcoholic Fatty Liver Disease as a Nexus of Metabolic and Hepatic Diseases. Cell Metab. 2018, 27, 22–41. [Google Scholar] [CrossRef] [Green Version]
  17. Chitturi, S.; Farrell, G.C. Etiopathogenesis of Nonalcoholic Steatohepatitis. Semin. Liver Dis. 2001, 21, 27–42. [Google Scholar] [CrossRef]
  18. Fromenty, B.; Pessayre, D. Impaired mitochondrial function in microvesicular steatosis effects of drugs, ethanol, hormones and cytokines. J. Hepatol. 1997, 26, 43–53. [Google Scholar] [CrossRef]
  19. Edmison, J.; McCullough, A.J. Pathogenesis of Non-alcoholic Steatohepatitis: Human Data. Clin. Liver Dis. 2007, 11, 75–104. [Google Scholar] [CrossRef]
  20. Day, C.P.; Saksena, S. Non-alcoholic steatohepatitis: Definitions and pathogenesis. J. Gastroenterol. Hepatol. 2002, 17, S377–S384. [Google Scholar] [CrossRef]
  21. Petersen, M.C.; Shulman, G.I. Mechanisms of Insulin Action and Insulin Resistance. Physiol. Rev. 2018, 98, 2133–2223. [Google Scholar] [CrossRef] [Green Version]
  22. Mantovani, A.; Byrne, C.D.; Bonora, E.; Targher, G. Nonalcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: A Meta-analysis. Diabetes Care 2018, 41, 372–382. [Google Scholar] [CrossRef] [Green Version]
  23. Byrne, C.D.; Targher, G. NAFLD as a driver of chronic kidney disease. J. Hepatol. 2020, 72, 785–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Cusi, K. Time to Include Nonalcoholic Steatohepatitis in the Management of Patients With Type 2 Diabetes. Diabetes Care 2020, 43, 275–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kleiner, D.E.; Brunt, E.M.; Wilson, L.A.; Behling, C.; Guy, C.; Contos, M.; Cummings, O.; Yeh, M.; Gill, R.; Chalasani, N.; et al. Association of Histologic Disease Activity With Progression of Nonalcoholic Fatty Liver Disease. JAMA Netw. Open 2019, 2, e1912565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sung, K.-C.; Wild, S.H.; Byrne, C.D. Resolution of Fatty Liver and Risk of Incident Diabetes. J. Clin. Endocrinol. Metab. 2013, 98, 3637–3643. [Google Scholar] [CrossRef] [PubMed]
  27. Preiss, D.; Sattar, N. Non-alcoholic fatty liver disease: An overview of prevalence, diagnosis, pathogenesis and treatment considerations. Clin. Sci. 2008, 115, 141–150. [Google Scholar] [CrossRef] [Green Version]
  28. Waxman, A. WHO’s global strategy on diet, physical activity and health: Response to a worldwide epidemic of non-communicable diseases. Scand. J. Nutr./Naringsforsk. 2004, 48, 58–60. [Google Scholar] [CrossRef]
  29. Perseghin, G.; Lattuada, G.; De Cobelli, F.; Ragogna, F.; Ntali, G.; Esposito, A.; Belloni, E.; Canu, T.; Terruzzi, I.; Scifo, P.; et al. Habitual Physical Activity Is Associated With Intrahepatic Fat Content in Humans. Diabetes Care 2007, 30, 683–688. [Google Scholar] [CrossRef] [Green Version]
  30. George, A.; Bauman, A.; Johnston, A.; Farrell, G.; Chey, T.; George, J. Independent effects of physical activity in patients with nonalcoholic fatty liver disease. Hepatology 2009, 50, 68–76. [Google Scholar] [CrossRef]
  31. Oh, S.; So, R.; Shida, T.; Matsuo, T.; Kim, B.; Akiyama, K.; Isobe, T.; Okamoto, Y.; Tanaka, K.; Shoda, J. High-Intensity Aerobic Exercise Improves Both Hepatic Fat Content and Stiffness in Sedentary Obese Men with Nonalcoholic Fatty Liver Disease. Sci. Rep. 2017, 7, srep43029. [Google Scholar] [CrossRef] [Green Version]
  32. Zhang, H.-J.; Pan, L.-L.; Ma, Z.-M.; Chen, Z.; Huang, Z.-F.; Sun, Q.; Lu, Y.; Han, C.-K.; Lin, M.-Z.; Li, X.-J.; et al. Long-term effect of exercise on improving fatty liver and cardiovascular risk factors in obese adults: A 1-year follow-up study. Diabetes Obes. Metab. 2016, 19, 284–289. [Google Scholar] [CrossRef]
  33. Jakovljevic, D.G.; Hallsworth, K.; Zalewski, P.; Thoma, C.; Klawe, J.J.; Day, C.P.; Newton, J.; Trenell, M.I. Resistance exercise improves autonomic regulation at rest and haemodynamic response to exercise in non-alcoholic fatty liver disease. Clin. Sci. 2013, 125, 143–149. [Google Scholar] [CrossRef] [Green Version]
  34. Pennisi, G.; Celsa, C.; Spatola, F.; Dallio, M.; Federico, A.; Petta, S. Pharmacological Therapy of Non-Alcoholic Fatty Liver Disease: What Drugs Are Available Now and Future Perspectives. Int. J. Environ. Public Health 2019, 16, 4334. [Google Scholar] [CrossRef] [Green Version]
  35. Golabi, P.; Locklear, C.T.; Austin, P.; Afdhal, S.; Byrns, M.; Gerber, L.; Younossi, Z.M. Effectiveness of exercise in hepatic fat mobilization in non-alcoholic fatty liver disease: Systematic review. World J. Gastroenterol. 2016, 22, 6318–6327. [Google Scholar] [CrossRef]
  36. Lee, I.M.; Shiroma, E.J.; Lobelo, F.; Puska, P.; Blair, S.N.; Katzmarzyk, P.T.; Lancet Physical Activity Series Working Group. Effect of physical inactivity on major non-communicable diseases worldwide: An analysis of burden of disease and life expectancy. Lancet 2012, 380, 219–229. [Google Scholar] [CrossRef] [Green Version]
  37. Goodpaster, B.H.; DeLany, J.P.; Otto, A.D.; Kuller, L.; Vockley, J.; South-Paul, J.E.; Thomas, S.B.; Brown, J.; McTigue, K.; Hames, K.V.; et al. Effects of diet and physical activity interventions on weight loss and cardiometabolic risk factors in severely obese adults: A randomized trial. JAMA 2010, 304, 1795–1802. [Google Scholar] [CrossRef] [Green Version]
  38. Finucane, F.M.; Sharp, S.J.; Purslow, L.R.; Horton, K.; Horton, J.; Savage, D.B.; Brage, S.; Besson, H.; Rolfe, E.D.L.; Sleigh, A.; et al. The effects of aerobic exercise on metabolic risk, insulin sensitivity and intrahepatic lipid in healthy older people from the Hertfordshire Cohort Study: A randomised controlled trial. Diabetologia 2010, 53, 624–631. [Google Scholar] [CrossRef] [Green Version]
  39. Slentz, C.A.; Bateman, L.A.; Willis, L.H.; Shields, A.T.; Tanner, C.J.; Piner, L.W.; Hawk, V.H.; Muehlbauer, M.J.; Samsa, G.P.; Nelson, R.C.; et al. Effects of aerobic vs. resistance training on visceral and liver fat stores, liver enzymes, and insulin resistance by HOMA in overweight adults from STRRIDE AT/RT. Am. J. Physiol. Metab. 2011, 301, E1033–E1039. [Google Scholar] [CrossRef] [Green Version]
  40. Wong, V.W.S.; Chan, R.S.M.; Wong, G.L.-H.; Cheung, B.H.K.; Chu, W.C.W.; Yeung, D.K.W.; Chim, A.M.L.; Lai, J.W.Y.; Li, L.S.; Sea, M.M.M.; et al. Community-based lifestyle modification programme for non-alcoholic fatty liver disease: A randomized controlled trial. J. Hepatol. 2013, 59, 536–542. [Google Scholar] [CrossRef]
  41. Zelber-Sagi, S.; Buch, A.; Yeshua, H.; Vaisman, N.; Webb, M.; Harari, G.; Kis, O.; Fliss-Isakov, N.; Izkhakov, E.; Halpern, Z.; et al. Effect of resistance training on non-alcoholic fatty-liver disease a randomized-clinical trial. World J. Gastroenterol. 2014, 20, 4382–4392. [Google Scholar] [CrossRef]
  42. Balducci, S.; Cardelli, P.; Pugliese, L.; D’Errico, V.; Haxhi, J.; Alessi, E.; Iacobini, C.; Menini, S.; Bollanti, L.; Conti, F.G.; et al. Volume-dependent effect of supervised exercise training on fatty liver and visceral adiposity index in subjects with type 2 diabetes The Italian Diabetes Exercise Study (IDES). Diabetes Res. Clin. Pract. 2015, 109, 355–363. [Google Scholar] [CrossRef]
  43. Cuthbertson, D.; Shojaee-Moradie, F.; Sprung, V.S.; Jones, H.; Pugh, C.J.; Richardson, P.; Kemp, G.; Barrett, M.; Jackson, N.C.; Thomas, E.; et al. Dissociation between exercise-induced reduction in liver fat and changes in hepatic and peripheral glucose homoeostasis in obese patients with non-alcoholic fatty liver disease. Clin. Sci. 2015, 130, 93–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Skrypnik, D.; Ratajczak, M.; Karolkiewicz, J.; Mądry, E.; Pupek-Musialik, D.; Hansdorfer-Korzon, R.; Walkowiak, J.; Jakubowski, H.; Bogdański, P. Effects of endurance and endurance–strength exercise on biochemical parameters of liver function in women with abdominal obesity. Biomed. Pharmacother. 2016, 80, 1–7. [Google Scholar] [CrossRef] [PubMed]
  45. Farzanegi, P.; Abbaszadeh, H.; Daloii, A.A.; Kazemi, M.; Sabbaghian, M.; Shoeibi, A.; Nabipour, R.; Abuhosseini, Z.; Azarbayjani, M.A. Effects of aerobic exercise on histopathology and toxicology of ZnO and nano ZnO in male rats. Toxicol. Environ. Chem. 2018, 100, 103–114. [Google Scholar] [CrossRef]
  46. Petroni, M.L.; Brodosi, L.; Bugianesi, E.; Marchesini, G. Management of non-alcoholic fatty liver disease. BMJ 2021, 372, m4747. [Google Scholar] [CrossRef] [PubMed]
  47. Romero-Gómez, M.; Zelber-Sagi, S.; Trenell, M. Treatment of NAFLD with diet, physical activity and exercise. J. Hepatol. 2017, 67, 829–846. [Google Scholar] [CrossRef] [Green Version]
  48. Chalasani, N.; Younossi, Z.; LaVine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef] [Green Version]
  49. Glen, J.; Floros, L.; Day, C.; Pryke, R. Non-alcoholic fatty liver disease (NAFLD): Summary of NICE guidance. BMJ 2016, 354, i4428. [Google Scholar] [CrossRef]
  50. Anderson, R.M.; Weindruch, R. Metabolic reprogramming, caloric restriction and aging. Trends Endocrinol. Metab. 2010, 21, 134–141. [Google Scholar] [CrossRef] [Green Version]
  51. Ludwig, D.S.; Ebbeling, C.B. The Carbohydrate-Insulin Model of Obesity: Beyond “Calories In, Calories Out”. JAMA Intern. Med. 2018, 178, 1098–1103. [Google Scholar] [CrossRef]
  52. Holmer, M.; Lindqvist, C.; Petersson, S.; Moshtaghi-Svensson, J.; Tillander, V.; Brismar, T.B.; Hagström, H.; Stål, P. Treatment of NAFLD with intermittent calorie restriction or low-carb high-fat diet—A randomised controlled trial. JHEP Rep. 2021, 3, 100256. [Google Scholar] [CrossRef]
  53. Vilar-Gomez, E.; Martinez-Perez, Y.; Calzadilla-Bertot, L.; Torres-Gonzalez, A.; Gra-Oramas, B.; Gonzalez-Fabian, L.; Friedman, S.L.; Diago, M.; Romero-Gomez, M. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology 2015, 149, 367–378.e5. [Google Scholar] [CrossRef]
  54. Kirk, E.; Reeds, D.N.; Finck, B.N.; Mayurranjan, M.S.; Patterson, B.W.; Klein, S. Dietary Fat and Carbohydrates Differentially Alter Insulin Sensitivity During Caloric Restriction. Gastroenterology 2009, 136, 1552–1560. [Google Scholar] [CrossRef] [Green Version]
  55. 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. [Google Scholar] [CrossRef]
  56. Musso, G.; Gambino, R.; De Michieli, F.; Cassader, M.; Rizzetto, M.; Durazzo, M.; Fagà, E.; Silli, B.; Pagano, G. Dietary habits and their relations to insulin resistance and postprandial lipemia in nonalcoholic steatohepatitis. Hepatology 2003, 37, 909–916. [Google Scholar] [CrossRef]
  57. Perdomo, C.M.; Frühbeck, G.; Escalada, J. Impact of Nutritional Changes on Nonalcoholic Fatty Liver Disease. Nutrients 2019, 11, 677. [Google Scholar] [CrossRef] [Green Version]
  58. Abdelmalek, M.F.; Suzuki, A.; Guy, C.; Unalp-Arida, A.; Colvin, R.; Johnson, R.J.; Diehl, A.M.; Nonalcoholic Steatohepatitis Clinical Research Network. Increased fructose consumption is associated with fibrosis severity in patients with nonalcoholic fatty liver disease. Hepatology 2010, 51, 1961–1971. [Google Scholar] [CrossRef] [Green Version]
  59. Gupta, V.; Mah, X.-J.; Garcia, M.C.; Antonypillai, C.; Van Der Poorten, D. Oily fish, coffee and walnuts: Dietary treatment for nonalcoholic fatty liver disease. World J. Gastroenterol. 2015, 21, 10621–10635. [Google Scholar] [CrossRef]
  60. Halima, B.H.; Sarra, K.; Houda, B.J.; Sonia, G.; Abdallah, A. Antidiabetic and Antioxidant Effects of Apple Cider Vinegar on Normal and Streptozotocin-Induced Diabetic Rats. Int. J. Vitam. Nutr. Res. 2018, 88, 223–233. [Google Scholar] [CrossRef]
  61. Fushimi, T.; Suruga, K.; Oshima, Y.; Fukiharu, M.; Tsukamoto, Y.; Goda, T. Dietary acetic acid reduces serum cholesterol and triacylglycerols in rats fed a cholesterol-rich diet. Br. J. Nutr. 2006, 95, 916–924. [Google Scholar] [CrossRef] [Green Version]
  62. Shishehbor, F.; Mansoori, A.; Sarkaki, A.; Jalali, M.; Latifi, S. Apple Cider Vinegar Attenuates Lipid Profile in Normal and Diabetic Rats. Pak. J. Biol. Sci. 2008, 11, 2634–2638. [Google Scholar] [CrossRef] [Green Version]
  63. Hadi, A.; Pourmasoumi, M.; Najafgholizadeh, A.; Clark, C.C.T.; Esmaillzadeh, A. The effect of apple cider vinegar on lipid profiles and glycemic parameters: A systematic review and meta-analysis of randomized clinical trials. BMC Complement. Med. Ther. 2021, 21, 179. [Google Scholar] [CrossRef] [PubMed]
  64. Ousaaid, D.; Laaroussi, H.; Bakour, M.; ElGhouizi, A.; Aboulghazi, A.; Lyoussi, B.; Elarabi, I. Beneficial Effects of Apple Vinegar on Hyperglycemia and Hyperlipidemia in Hypercaloric-Fed Rats. J. Diabetes Res. 2020, 2020, 9284987. [Google Scholar] [CrossRef] [PubMed]
  65. Haufe, S.; Engeli, S.; Kast, P.; Böhnke, J.; Utz, W.; Haas, V.; Hermsdorf, M.; Mähler, A.; Wiesner, S.; Birkenfeld, A.L.; et al. Randomized comparison of reduced fat and reduced carbohydrate hypocaloric diets on intrahepatic fat in overweight and obese human subjects. Hepatology 2011, 53, 1504–1514. [Google Scholar] [CrossRef] [PubMed]
  66. Browning, J.D.; Baker, J.A.; Rogers, T.; Davis, J.; Satapati, S.; Burgess, S.C. Short-term weight loss and hepatic triglyceride reduction: Evidence of a metabolic advantage with dietary carbohydrate restriction. Am. J. Clin. Nutr. 2011, 93, 1048–1052. [Google Scholar] [CrossRef] [Green Version]
  67. Ryan, M.C.; Itsiopoulos, C.; Thodis, T.; Ward, G.; Trost, N.; Hofferberth, S.; O’Dea, K.; Desmond, P.V.; Johnson, N.A.; Wilson, A.M. The Mediterranean diet improves hepatic steatosis and insulin sensitivity in individuals with non-alcoholic fatty liver disease. J. Hepatol. 2013, 59, 138–143. [Google Scholar] [CrossRef]
  68. Misciagna, G.; Del Pilar Diaz, M.; Caramia, D.V.; Bonfiglio, C.; Franco, I.; Noviello, M.R.; Chiloiro, M.; Abbrescia, D.I.; Mirizzi, A.; Tanzi, M.; et al. Effect of a low glycemic index Mediterranean diet on non-alcoholic fatty liver disease. A randomized controlled clinici trial. J. Nutr. Health Aging 2017, 21, 404–412. [Google Scholar] [CrossRef]
  69. Abenavoli, L.; Greco, M.; Milic, N.; Accattato, F.; Foti, D.; Gulletta, E.; Luzza, F. Effect of Mediterranean Diet and Antioxidant Formulation in Non-Alcoholic Fatty Liver Disease: A Randomized Study. Nutrients 2017, 9, 870. [Google Scholar] [CrossRef]
  70. Markova, M.; Pivovarova, O.R.; Hornemann, S.; Sucher, S.; Frahnow, T.; Wegner, K.; Machann, J.; Petzke, K.J.; Hierholzer, J.; Lichtinghagen, R.; et al. Isocaloric Diets High in Animal or Plant Protein Reduce Liver Fat and Inflammation in Individuals With Type 2 Diabetes. Gastroenterology 2017, 152, 571–585.e8. [Google Scholar] [CrossRef] [Green Version]
  71. Katsagoni, C.N.; Papatheodoridis, G.V.; Ioannidou, P.; Deutsch, M.; Alexopoulou, A.; Papadopoulos, N.; Papageorgiou, M.-V.; Fragopoulou, E.; Kontogianni, M. Improvements in clinical characteristics of patients with non-alcoholic fatty liver disease, after an intervention based on the Mediterranean lifestyle: A randomised controlled clinical trial. Br. J. Nutr. 2018, 120, 164–175. [Google Scholar] [CrossRef]
  72. Marin-Alejandre, B.A.; Abete, I.; Cantero, I.; Monreal, J.I.; Elorz, M.; Herrero, J.I.; Benito-Boillos, A.; Quiroga, J.; Martinez-Echeverria, A.; Uriz-Otano, J.I.; et al. The Metabolic and Hepatic Impact of Two Personalized Dietary Strategies in Subjects with Obesity and Nonalcoholic Fatty Liver Disease: The Fatty Liver in Obesity (FLiO) Randomized Controlled Trial. Nutrients 2019, 11, 2543. [Google Scholar] [CrossRef] [Green Version]
  73. Gepner, Y.; Shelef, I.; Komy, O.; Cohen, N.; Schwarzfuchs, D.; Bril, N.; Rein, M.; Serfaty, D.; Kenigsbuch, S.; Zelicha, H.; et al. The beneficial effects of Mediterranean diet over low-fat diet may be mediated by decreasing hepatic fat content. J. Hepatol. 2019, 71, 379–388. [Google Scholar] [CrossRef] [Green Version]
  74. Meir, A.Y.; Rinott, E.; Tsaban, G.; Zelicha, H.; Kaplan, A.; Rosen, P.; Shelef, I.; Youngster, I.; Shalev, A.; Blüher, M.; et al. Effect of green-Mediterranean diet on intrahepatic fat: The DIRECT PLUS randomised controlled trial. Gut 2021, 70, 2085–2095. [Google Scholar] [CrossRef]
  75. Xu, C.; Markova, M.; Seebeck, N.; Loft, A.; Hornemann, S.; Gantert, T.; Kabisch, S.; Herz, K.; Loske, J.; Ost, M.; et al. High-protein diet more effectively reduces hepatic fat than low-protein diet despite lower autophagy and FGF21 levels. Liver Int. 2020, 40, 2982–2997. [Google Scholar] [CrossRef]
  76. Goss, A.M.; Dowla, S.; Pendergrass, M.; Ashraf, A.; Bolding, M.; Morrison, S.; Amerson, A.; Soleymani, T.; Gower, B. Effects of a carbohydrate-restricted diet on hepatic lipid content in adolescents with non-alcoholic fatty liver disease: A pilot, randomized trial. Pediatr. Obes. 2020, 15, e12630. [Google Scholar] [CrossRef]
  77. Brunt, E.M.; Wong, V.W.S.; Nobili, V.; Day, C.P.; Sookoian, S.; Maher, J.J.; Bugianesi, E.; Sirlin, C.B.; Neuschwander-Tetri, B.A.; Rinella, M.E. Nonalcoholic fatty liver disease. Nat. Rev. Dis. Prim. 2015, 1, 15080. [Google Scholar] [CrossRef]
  78. Jiang, X.; Zheng, J.; Zhang, S.; Wang, B.; Wu, C.; Guo, X. Advances in the Involvement of Gut Microbiota in Pathophysiology of NAFLD. Front. Med. 2020, 7, 361. [Google Scholar] [CrossRef]
  79. Burguera, B.; Agusti, A.; Arner, P.; Baltasar, A.; Barbe, F.; Barcelo, A.; Breton, I.; Cabanes, T.; Casanueva, F.F.; Couce, M.E.; et al. Critical assessment of the current guidelines for the management and treatment of morbidly obese patients. J. Endocrinol. Investig. 2007, 30, 844–852. [Google Scholar] [CrossRef]
  80. Nguyen, N.T.; Varela, J.E. Bariatric surgery for obesity and metabolic disorders: State of the art. Nat. Rev. Gastroenterol. Hepatol. 2016, 14, 160–169. [Google Scholar] [CrossRef]
  81. Alghamdi, S.; Mirghani, H.; Alhazmi, K.; Alatawi, A.M.; Brnawi, H.; Alrasheed, T.; Badoghaish, W. Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy effects on obesity comorbidities: A systematic review and meta-analysis. Front. Surg. 2022, 9, 953804. [Google Scholar] [CrossRef]
  82. Seeras, K.; Lopez, P.P. Sleeve Gastrectomy; Starpearls Pubilshing: Treasure Island, FL, USA, 2021. [Google Scholar]
  83. Lee, Y.; Doumouras, A.G.; Yu, J.; Brar, K.; Banfield, L.; Gmora, S.; Anvari, M.; Hong, D. Complete Resolution of Nonalcoholic Fatty Liver Disease After Bariatric Surgery: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2019, 17, 1040–1060.e11. [Google Scholar] [CrossRef] [Green Version]
  84. Schwenger, K.J.; Fischer, S.E.; Jackson, T.; Okrainec, A.; Allard, J.P. In nonalcoholic fatty liver disease, Roux-en-Y gastric bypass improves liver histology while persistent disease is associated with lower improvements in waist circumference and glycemic control. Surg. Obes. Relat. Dis. 2018, 14, 1233–1239. [Google Scholar] [CrossRef]
  85. Weiner, R. Surgical Treatment of Non-Alcoholic Steatohepatitis and Non-Alcoholic Fatty Liver Disease. Dig. Dis. 2010, 28, 274–279. [Google Scholar] [CrossRef] [PubMed]
  86. Moretto, M.; Kupski, C.; Da Silva, V.D.; Padoin, A.; Mottin, C.C. Effect of Bariatric Surgery on Liver Fibrosis. Obes. Surg. 2011, 22, 1044–1049. [Google Scholar] [CrossRef] [PubMed]
  87. Tai, C.-M.; Huang, C.-K.; Hwang, J.-C.; Chiang, H.; Chang, C.-Y.; Lee, C.-T.; Yu, M.-L.; Lin, J.-T. Improvement of Nonalcoholic Fatty Liver Disease After Bariatric Surgery in Morbidly Obese Chinese Patients. Obes. Surg. 2011, 22, 1016–1021. [Google Scholar] [CrossRef] [PubMed]
  88. Vargas, H.A.C.; Allende, H.; Lecube, A.; Salcedo, M.T.; Baena-Fustegueras, A.J.; Fort, J.M.; Rivero, R.F.N.; Ferrer, R.; Catalán, R.; Pardina, E.; et al. Surgically induced weight loss by gastric bypass improves non alcoholic fatty liver disease in morbid obese patients. World J. Hepatol. 2012, 4, 382–388. [Google Scholar] [CrossRef] [Green Version]
  89. Taitano, A.A.; Markow, M.; Finan, J.E.; Wheeler, D.E.; Gonzalvo, J.P.; Murr, M.M. Bariatric surgery improves histological features of nonalcoholic fatty liver disease and liver fibrosis. J. Gastrointest. Surg. 2015, 19, 429–437. [Google Scholar] [CrossRef]
  90. Lassailly, G.; Caiazzo, R.; Buob, D.; Pigeyre, M.; Verkindt, H.; Labreuche, J.; Raverdy, V.; Leteurtre, E.; Dharancy, S.; Louvet, A.; et al. Bariatric Surgery Reduces Features of Nonalcoholic Steatohepatitis in Morbidly Obese Patients. Gastroenterology 2015, 149, 379–388, quiz e315–e376. [Google Scholar] [CrossRef] [Green Version]
  91. Aldoheyan, T.; Hassanain, M.; Al-Mulhim, A.; Al-Sabhan, A.; Al-Amro, S.; Bamehriz, F.; Al-Khalidi, H. The effects of bariatric surgeries on nonalcoholic fatty liver disease. Surg. Endosc. 2017, 31, 1142–1147. [Google Scholar] [CrossRef]
  92. Parker, B.M.; Wu, J.; You, J.; Barnes, D.S.; Yerian, L.; Kirwan, J.P.; Schauer, P.R.; Sessler, D.I. Reversal of fibrosis in patients with nonalcoholic steatohepatosis after gastric bypass surgery. BMC Obes. 2017, 4, 32. [Google Scholar] [CrossRef] [Green Version]
  93. Esquivel, C.M.; Garcia, M.; Armando, L.; Ortiz, G.; Lascano, F.M.; Foscarini, J.M. Laparoscopic Sleeve Gastrectomy Resolves NAFLD: Another Formal Indication for Bariatric Surgery? Obes. Surg. 2018, 28, 4022–4033. [Google Scholar] [CrossRef]
  94. Pooler, B.D.; Wiens, C.N.; McMillan, A.; Artz, N.S.; Schlein, A.; Covarrubias, Y.; Hooker, J.; Schwimmer, J.B.; Funk, L.M.; Campos, G.M.; et al. Monitoring Fatty Liver Disease with MRI Following Bariatric Surgery: A Prospective, Dual-Center Study. Radiology 2019, 290, 682–690. [Google Scholar] [CrossRef]
  95. Mantovani, A.; Byrne, C.; Scorletti, E.; Mantzoros, C.; Targher, G. Efficacy and safety of anti-hyperglycaemic drugs in patients with non-alcoholic fatty liver disease with or without diabetes: An updated systematic review of randomized controlled trials. Diabetes Metab. 2020, 46, 427–441. [Google Scholar] [CrossRef]
  96. Kalavalapalli, S.; Bril, F.; Guingab, J.; Vergara, A.; Garrett, T.J.; Sunny, E.N.; Cusi, K. Impact of exenatide on mitochondrial lipid metabolism in mice with nonalcoholic steatohepatitis. J. Endocrinol. 2019, 241, 293–305. [Google Scholar] [CrossRef]
  97. Katsiki, N.; Perakakis, N.; Mantzoros, C. Effects of sodium-glucose co-transporter-2 (SGLT2) inhibitors on non-alcoholic fatty liver disease/non-alcoholic steatohepatitis: Ex quo et quo vadimus? Metabolism 2019, 98, 3–9. [Google Scholar] [CrossRef]
  98. Mayerson, A.B.; Hundal, R.S.; Dufour, S.; Lebon, V.; Befroy, D.; Cline, G.W.; Enocksson, S.; Inzucchi, S.E.; Shulman, G.I.; Petersen, K.F. The Effects of Rosiglitazone on Insulin Sensitivity, Lipolysis, and Hepatic and Skeletal Muscle Triglyceride Content in Patients With Type 2 Diabetes. Diabetes 2002, 51, 797–802. [Google Scholar] [CrossRef] [Green Version]
  99. Dos Coelho, F.S.; Borges-Canha, M.; von Hafe, M.; Neves, J.S.; Vale, C.; Leite, A.R.; Carvalho, D.; Leite-Moreira, A. Effects of sodium-glucose co-transporter 2 inhibitors on liver parameters and steatosis: A meta-analysis of randomized clinical trials. Diabetes Metab. Res. Rev. 2021, 37, e3413. [Google Scholar]
  100. Shibuya, T.; Fushimi, N.; Kawai, M.; Yoshida, Y.; Hachiya, H.; Ito, S.; Kawai, H.; Ohashi, N.; Mori, A. Luseogliflozin improves liver fat deposition compared to metformin in type 2 diabetes patients with non-alcoholic fatty liver disease: A prospective randomized controlled pilot study. Diabetes Obes. Metab. 2017, 20, 438–442. [Google Scholar] [CrossRef]
  101. Eriksson, J.W.; Lundkvist, P.; Jansson, P.-A.; Johansson, L.; Kvarnström, M.; Moris, L.; Miliotis, T.; Forsberg, G.-B.; Risérus, U.; Lind, 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. [Google Scholar] [CrossRef] [Green Version]
  102. Sattar, N.; Fitchett, D.; Hantel, S.; George, J.T.; Zinman, B. Empagliflozin is associated with improvements in liver enzymes potentially consistent with reductions in liver fat: Results from randomised trials including the EMPA-REG OUTCOME® trial. Diabetologia 2018, 61, 2155–2163. [Google Scholar] [CrossRef] [Green Version]
  103. Gross, B.; Pawlak, M.; Lefebvre, P.; Staels, B. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nat. Rev. Endocrinol. 2017, 13, 36–49. [Google Scholar] [CrossRef]
  104. Belfort, R.; Harrison, S.A.; Brown, K.; Darland, C.; Finch, J.; Hardies, J.; Balas, B.; Gastaldelli, A.; Tio, F.; Pulcini, J.; et al. A Placebo-Controlled Trial of Pioglitazone in Subjects with Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2006, 355, 2297–2307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Boettcher, E.; Csako, G.; Pucino, F.; Wesley, R.; Loomba, R. Meta-analysis: Pioglitazone improves liver histology and fibrosis in patients with non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 2011, 35, 66–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Al-Mrabeh, A.; Hollingsworth, K.G.; Shaw, J.A.M.; McConnachie, A.; Sattar, N.; Lean, M.E.J.; Taylor, R. 2-year remission of type 2 diabetes and pancreas morphology: A post-hoc analysis of the DiRECT open-label, cluster-randomised trial. Lancet Diabetes Endocrinol. 2020, 8, 939–948. [Google Scholar] [CrossRef] [PubMed]
  107. Thomas, M.K.; Nikooienejad, A.; Bray, R.; Cui, X.; Wilson, J.; Duffin, K.; Milicevic, Z.; Haupt, A.; Robins, A.D. Dual GIP and GLP-1 Receptor Agonist Tirzepatide Improves Beta-cell Function and Insulin Sensitivity in Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2020, 106, 388–396. [Google Scholar] [CrossRef]
  108. Dutour, A.; Abdesselam, I.; Ancel, P.; Kober, F.; Mrad, G.; Darmon, P.; Ronsin, O.; Pradel, V.; Lesavre, N.; Martin, J.C.; et al. Exenatide decreases liver fat content and epicardial adipose tissue in patients with obesity and type 2 diabetes: A prospective randomized clinical trial using magnetic resonance imaging and spectroscopy. Diabetes Obes. Metab. 2016, 18, 882–891. [Google Scholar] [CrossRef]
  109. Armstrong, M.J.; Houlihan, D.D.; Rowe, I.A.; Clausen, W.H.O.; Elbrønd, B.; Gough, S.C.L.; Tomlinson, J.W.; Newsome, P.N. Safety and efficacy of liraglutide in patients with type 2 diabetes and elevated liver enzymes: Individual patient data meta-analysis of the LEAD program. Aliment. Pharmacol. Ther. 2012, 37, 234–242. [Google Scholar] [CrossRef]
  110. 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. 2015, 64, 399–408. [Google Scholar] [CrossRef] [Green Version]
  111. Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; Abouda, G.; Aldersley, A.M.; et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 2016, 387, 679–690. [Google Scholar] [CrossRef] [Green Version]
  112. Newsome, P.; Francque, S.; Harrison, S.; Ratziu, V.; Van Gaal, L.; Calanna, S.; Hansen, M.; Linder, M.; Sanyal, A. Effect of semaglutide on liver enzymes and markers of inflammation in subjects with type 2 diabetes and/or obesity. Aliment. Pharmacol. Ther. 2019, 50, 193–203. [Google Scholar] [CrossRef] [Green Version]
  113. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.-S.; Harrison, S.A. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124. [Google Scholar] [CrossRef]
  114. Cui, J.; Philo, L.; Nguyen, P.; Hofflich, H.; Hernandez, C.; Bettencourt, R.; Richards, L.; Salotti, J.; Bhatt, A.; Hooker, J.; et al. Sitagliptin vs. placebo for non-alcoholic fatty liver disease: A randomized controlled trial. J. Hepatol. 2016, 65, 369–376. [Google Scholar] [CrossRef] [Green Version]
  115. Joy, T.R.; McKenzie, A.C.; Tirona, R.G.; Summers, K.; Seney, S.; Chakrabarti, S.; Malhotra, N.; Beaton, M.D. Sitagliptin in patients with non-alcoholic steatohepatitis: A randomized, placebo-controlled trial. World J. Gastroenterol. 2017, 23, 141–150. [Google Scholar] [CrossRef]
  116. Neuschwander-Tetri, B.A.; Brunt, E.M.; Wehmeier, K.R.; Oliver, D.; Bacon, B.R. Improved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-γ ligand rosiglitazone. Hepatology 2003, 38, 1008–1017. [Google Scholar] [CrossRef]
  117. Ratziu, V.; Giral, P.; Jacqueminet, S.; Charlotte, F.; Hartemann–Heurtier, A.; Serfaty, L.; Podevin, P.; Lacorte, J.M.; Bernhardt, C.; Bruckert, E.; et al. Rosiglitazone for Nonalcoholic Steatohepatitis: One-Year Results of the Randomized Placebo-Controlled Fatty Liver Improvement With Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology 2008, 135, 100–110. [Google Scholar] [CrossRef]
  118. Cusi, K.; Orsak, B.; Bril, F.; Lomonaco, R.; Hecht, J.; Ortiz-Lopez, C.; Tio, F.; Hardies, J.; Darland, C.; Musi, N.; et al. Long-Term Pioglitazone Treatment for Patients With Nonalcoholic Steatohepatitis and Prediabetes or Type 2 Diabetes Mellitus: A Randomized Trial. Ann. Intern. Med. 2016, 165, 305–315. [Google Scholar] [CrossRef]
  119. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef] [Green Version]
  120. Cusi, K.; Bril, F.; Barb, D.; Polidori, D.; Sha, S.; Ghosh, A.; Bs, K.F.; Sunny, N.E.; Kalavalapalli, S.; Pettus, J.; et al. Effect of canagliflozin treatment on hepatic triglyceride content and glucose metabolism in patients with type 2 diabetes. Diabetes Obes. Metab. 2018, 21, 812–821. [Google Scholar] [CrossRef]
  121. Latva-Rasku, A.; Honka, M.-J.; Kullberg, J.; Mononen, N.; Lehtimäki, T.; Saltevo, J.; Kirjavainen, A.K.; Saunavaara, V.; Iozzo, P.; Johansson, L.; et al. The SGLT2 Inhibitor Dapagliflozin Reduces Liver Fat but Does Not Affect Tissue Insulin Sensitivity: A Randomized, Double-Blind, Placebo-Controlled Study With 8-Week Treatment in Type 2 Diabetes Patients. Diabetes Care 2019, 42, 931–937. [Google Scholar] [CrossRef] [Green Version]
  122. Shimizu, M.; Suzuki, K.; Kato, K.; Jojima, T.; Iijima, T.; Murohisa, T.; Iijima, M.; Takekawa, H.; Usui, I.; Hiraishi, H.; 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 Obes. Metab. 2018, 21, 285–292. [Google Scholar] [CrossRef]
  123. Kahl, S.; Gancheva, S.; Straßburger, K.; Herder, C.; Machann, J.; Katsuyama, H.; Kabisch, S.; Henkel, E.; Kopf, S.; Lagerpusch, M.; et al. Empagliflozin Effectively Lowers Liver Fat Content in Well-Controlled Type 2 Diabetes: A Randomized, Double-Blind, Phase 4, Placebo-Controlled Trial. Diabetes Care 2019, 43, 298–305. [Google Scholar] [CrossRef] [Green Version]
  124. Kuchay, M.S.; Krishan, S.; Mishra, S.K.; Farooqui, K.J.; Singh, M.K.; Wasir, J.S.; Bansal, B.; Kaur, P.; Jevalikar, G.; Gill, H.K.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Jeon, Y.; Son, K.Y. Effects of different definitions of low muscle mass on its association with metabolic syndrome in older adults: A Korean nationwide study. Geriatr. Gerontol. Int. 2021, 21, 1003–1009. [Google Scholar] [CrossRef] [PubMed]
  127. Pose, E.; Trebicka, J.; Mookerjee, R.P.; Angeli, P.; Ginès, P. Statins: Old drugs as new therapy for liver diseases? J. Hepatol. 2019, 70, 194–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Dongiovanni, P.; Petta, S.; Mannisto, V.; Mancina, R.M.; Pipitone, R.; Karja, V.; Maggioni, M.; Kakela, P.; Wiklund, O.; Mozzi, E.; et al. Statin use and non-alcoholic steatohepatitis in at risk individuals. J. Hepatol. 2015, 63, 705–712. [Google Scholar] [CrossRef]
  129. Lee, J.; Lee, H.W.; Lee, K.S.; Lee, H.S.; Park, J.Y. Effects of Statin Use on the Development and Progression of Nonalcoholic Fatty Liver Disease: A Nationwide Nested Case-Control Study. Am. J. Gastroenterol. 2021, 116, 116–124. [Google Scholar] [CrossRef]
  130. Nakade, Y.; Murotani, K.; Inoue, T.; Kobayashi, Y.; Yamamoto, T.; Ishii, N.; Ohashi, T.; Ito, K.; Fukuzawa, Y.; Yoneda, M. Ezetimibe for the treatment of non-alcoholic fatty liver disease: A meta-analysis. Hepatol. Res. 2017, 47, 1417–1428. [Google Scholar] [CrossRef]
  131. Dewidar, B.; Kahl, S.; Pafili, K.; Roden, M. Metabolic liver disease in diabetes—From mechanisms to clinical trials. Metabolism 2020, 111, 154299. [Google Scholar] [CrossRef]
  132. Lee, C.-H.; Fu, Y.; Yang, S.-J.; Chi, C.-C. Effects of Omega-3 Polyunsaturated Fatty Acid Supplementation on Non-Alcoholic Fatty Liver: A Systematic Review and Meta-Analysis. Nutrients 2020, 12, 2769. [Google Scholar] [CrossRef]
  133. Valenzuela, R.; Videla, L.A. Impact of the Co-Administration of N-3 Fatty Acids and Olive Oil Components in Preclinical Nonalcoholic Fatty Liver Disease Models: A Mechanistic View. Nutrients 2020, 12, 499. [Google Scholar] [CrossRef] [Green Version]
  134. Huang, T.; Wu, T.; Wu, Y.; Li, X.; Tan, J.; Shen, C.; Xiong, S.; Feng, Z.; Gao, S.; Li, H.; et al. Long-term statins administration exacerbates diabetic nephropathy via ectopic fat deposition in diabetic mice. Nat. Commun. 2023, 14, 390. [Google Scholar] [CrossRef]
  135. Mansi, I.A.; Chansard, M.; Lingvay, I.; Zhang, S.; Halm, E.A.; Alvarez, C.A. Association of Statin Therapy Initiation With Diabetes Progression: A Retrospective Matched-Cohort Study. JAMA Intern. Med. 2021, 181, 1562–1574. [Google Scholar] [CrossRef]
  136. Cariou, B.; Byrne, C.D.; Loomba, R.; Sanyal, A.J. Nonalcoholic fatty liver disease as a metabolic disease in humans: A literature review. Diabetes Obes. Metab. 2021, 23, 1069–1083. [Google Scholar] [CrossRef]
  137. Kong, B.; Luyendyk, J.P.; Tawfik, O.; Guo, G.L. Farnesoid X Receptor Deficiency Induces Nonalcoholic Steatohepatitis in Low-Density Lipoprotein Receptor-Knockout Mice Fed a High-Fat Diet. Experiment 2008, 328, 116–122. [Google Scholar] [CrossRef] [Green Version]
  138. Younossi, Z.M.; Ratziu, V.; Loomba, R.; Rinella, M.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; Geier, A.; Beckebaum, S.; Newsome, P.N.; 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. [Google Scholar] [CrossRef] [Green Version]
  139. Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Van Natta, M.L.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; 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. [Google Scholar] [CrossRef] [Green Version]
  140. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef]
  141. Miyazaki, M.; Flowers, M.T.; Sampath, H.; Chu, K.; Otzelberger, C.; Liu, X.; Ntambi, J.M. Hepatic stearoyl-CoA desaturase-1 deficiency protects mice from carbohydrate-induced adiposity and hepatic steatosis. Cell Metab. 2007, 6, 484–496. [Google Scholar] [CrossRef] [Green Version]
  142. One-Year Results of the Global Phase 2b Randomized Placebo-Controlled ARREST Trial of Aramchol, a Stearoyl CoA Desaturasemodulator in NASH Patients. Available online: https://www.natap.org/2018/AASLD/AASLD_222.htm (accessed on 1 November 2018).
  143. Garvey, W.T.; Mechanick, J.I.; Brett, E.M.; Garber, A.J.; Hurley, D.L.; Jastreboff, A.M.; Nadolsky, K.; Pessah-Pollack, R.; Plodkowski, R. American association of clinical endocrinologists and american college of endocrinology comprehensive clinical practice guidelines for medical care of patients with obesity. Endocr. Pract. 2016, 22 (Suppl. 3), 1–203. [Google Scholar] [CrossRef] [Green Version]
  144. Kotronen, A.; Seppänen-Laakso, T.; Westerbacka, J.; Kiviluoto, T.; Arola, J.; Ruskeepää, A.L.; Matej, O.; Hannele, Y.-J. Hepatic stearoyl-CoA desaturase (SCD)-1 activity and diacylglycerol but not ceramide concentrations are increased in the nonalcoholic human fatty liver. Diabetes 2009, 58, 203–208. [Google Scholar] [CrossRef] [Green Version]
  145. Gutiérrez-Juárez, R.; Pocai, A.; Mulas, C.; Ono, H.; Bhanot, S.; Monia, B.P.; Rossetti, L. Critical role of stearoyl-CoA desaturase-1 (SCD1) in the onset of diet-induced hepatic insulin resistance. J. Clin. Investig. 2006, 116, 1686–1695. [Google Scholar] [CrossRef] [PubMed]
  146. Khera, R.; Murad, M.H.; Chandar, A.K.; Dulai, P.S.; Wang, Z.; Prokop, L.J.; Loomba, R.; Camilleri, M.; Singh, S. Association of Pharmacological Treatments for Obesity With Weight Loss and Adverse Events: A Systematic Review and Meta-analysis. JAMA 2016, 315, 2424–2434. [Google Scholar] [CrossRef] [Green Version]
  147. Damci, T.; Yalin, S.; Balci, H.; Osar, Z.; Korugan, U.; Ozyazar, M.; Ilkova, H. Orlistat augments postprandial increases in glucagon-like peptide 1 in obese type 2 diabetic patients. Diabetes Care 2004, 27, 1077–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Ruof, J.; Golay, A.; Berne, C.; Collin, C.; Lentz, J.; Maetzel, A. Orlistat in responding obese type 2 diabetic patients: Meta-analysis findings and cost-effectiveness as rationales for reimbursement in Sweden and Switzerland. Int. J. Obes. 2005, 29, 517–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Pan, C.S.; Stanley, T.L. Effect of Weight Loss Medications on Hepatic Steatosis and Steatohepatitis: A Systematic Review. Front. Endocrinol. 2020, 11, 70. [Google Scholar] [CrossRef] [Green Version]
  150. Bailey, C.J.; Flatt, P.R.; Conlon, J.M. An update on peptide-based therapies for type 2 diabetes and obesity. Peptides 2023, 161, 170939. [Google Scholar] [CrossRef]
  151. Gastaldelli, A.; Cusi, K.; Landó, L.F.; Bray, R.; Brouwers, B.; Rodríguez, Á. Effect of tirzepatide versus insulin degludec on liver fat content and abdominal adipose tissue in people with type 2 diabetes (SURPASS-3 MRI): A substudy of the randomised, open-label, parallel-group, phase 3 SURPASS-3 trial. Lancet Diabetes Endocrinol. 2022, 10, 393–406. [Google Scholar] [CrossRef]
  152. Cuthbertson, D.J.; Irwin, A.; Gardner, C.J.; Daousi, C.; Purewal, T.; Furlong, N.; Goenka, N.; Thomas, E.L.; Adams, V.L.; Pushpakom, S.P.; et al. Improved Glycaemia Correlates with Liver Fat Reduction in Obese, Type 2 Diabetes, Patients Given Glucagon-Like Peptide-1 (GLP-1) Receptor Agonists. PLoS ONE 2012, 7, e50117. [Google Scholar] [CrossRef] [Green Version]
  153. Neeland, I.J.; Marso, S.P.; Ayers, C.R.; Lewis, B.; Oslica, R.; Francis, W.; Rodder, S.; Pandey, A.; Joshi, P.H. Effects of liraglutide on visceral and ectopic fat in adults with overweight and obesity at high cardiovascular risk: A randomised, double-blind, placebo-controlled, clinical trial. Lancet Diabetes Endocrinol. 2021, 9, 595–605. [Google Scholar] [CrossRef]
  154. Mantovani, A.; Petracca, G.; Beatrice, G.; Csermely, A.; Lonardo, A.; Targher, G. Glucagon-Like Peptide-1 Receptor Agonists for Treatment of Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis: An Updated Meta-Analysis of Randomized Controlled Trials. Metabolites 2021, 11, 73. [Google Scholar] [CrossRef]
  155. Hartman, M.L.; Sanyal, A.J.; Loomba, R.; Wilson, J.M.; Nikooienejad, A.; Bray, R.; Karanikas, C.A.; Duffin, K.L.; Robins, D.A.; Haupt, A. Effects of Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide on Biomarkers of Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes. Diabetes Care 2020, 43, 1352–1355. [Google Scholar] [CrossRef]
  156. Volpe, S.; Lisco, G.; Fanelli, M.; Racaniello, D.; Colaianni, V.; Triggiani, D.; Donghia, R.; Crudele, L.; Rinaldi, R.; Sabbà, C.; et al. Once-Weekly Subcutaneous Semaglutide Improves Fatty Liver Disease in Patients with Type 2 Diabetes: A 52-Week Prospective Real-Life Study. Nutrients 2022, 14, 4673. [Google Scholar] [CrossRef]
  157. Flint, A.; Andersen, G.; Hockings, P.; Johansson, L.; Morsing, A.; Palle, M.S.; Vogl, T.; Loomba, R.; Plum-Mörschel, L. Randomised clinical trial: Semaglutide versus placebo reduced liver steatosis but not liver stiffness in subjects with non-alcoholic fatty liver disease assessed by magnetic resonance imaging. Aliment. Pharmacol. Ther. 2021, 54, 1150–1161. [Google Scholar] [CrossRef]
  158. Mantovani, A.; Byrne, C.D.; 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. Lancet Gastroenterol. Hepatol. 2022, 7, 367–378. [Google Scholar] [CrossRef]
  159. Alkhouri, N.; Herring, R.; Kabler, H.; Kayali, Z.; Hassanein, T.; Kohli, A.; Huss, R.S.; Zhu, Y.; Billin, A.N.; Damgaard, L.H.; et al. Safety and efficacy of combination therapy with semaglutide, cilofexor and firsocostat in patients with non-alcoholic steatohepatitis: A randomised, open-label phase II trial. J. Hepatol. 2022, 77, 607–618. [Google Scholar] [CrossRef]
  160. Vieira Barbosa, J.; Lai, M. Nonalcoholic Fatty Liver Disease Screening in Type 2 Diabetes Mellitus Patients in the Primary Care Setting. Hepatol. Commun. 2020, 5, 158–167. [Google Scholar] [CrossRef]
Table 1. Effects of exercise in patients with NAFLD.
Table 1. Effects of exercise in patients with NAFLD.
StudyNumber of PatientsType of ExerciseDuration of InterventionOutcomes
Goodpaster et al. (2010) [37]n = 130Aerobic Exercise with diet6 months vs. 12 months↓ hepatic lipid content in 12 months, equal ↓ of insulin resistant
Finucane et al. (2010) [38]n = 100Aerobic Exercise vs. control12 weeks↓ hepatic lipid content
Slentz et al. (2011) [39]n = 196Aerobic Exercise vs. Resistance Exercise vs. Aerobic + Resistance Exercise8 monthsGroups including aerobic exercise: higher ↓ hepatic fat content, ↓ ALT, and ↓ insulin resistance
Jakovljevic et al. (2013) [33]n = 17Resistance training vs. Control8 weeks↓ hepatic fat contents in resistance training group
Wong et al. (2013) [40]n = 154Aerobic Exercise vs. Control12 months↓ hepatic fat content
Zelber-Sagi et al. (2014) [41]n = 82Resistance Exercise vs. Control12 weeksImproving of steatosis and inflammation
Balducci et al. (2015) [42]n = 606Aerobic Exercise + Resistance Exercise vs. Control12 months↓ fatty liver index
Cuthbertson et al. (2016) [43]n = 69Aerobic Exercise vs. Control16 weeks↓ hepatic lipid content, ↑peripheral insulin sensitivity
Zhang et al. (2016) [32]n = 220Aerobic Exercise vs. control12 months↓ hepatic fat content
Skrypnik et al. (2016) [44]n = 44Aerobic Exercise vs. Aerobic + Resistance Exercise3 monthsGreater reduction in ALT and AST
Oh et al. (2017) [31]n = 61Aerobic + Resistance training12 weeks↓ ALT, ↓ AST and ↓ TG
Farzanegi et al. (2018) [45]n = 49 (rats)Aerobic training4 weeks↓ hepatic cell apoptosis, ↓ ALT, ↓ AST and ↓ ALP
AST: aspartate aminotransferase, ALT: alanine aminotransferase, TG: triglycerides, ALP: alkaline phosphatase, ↓: decrease.
Table 3. The effectiveness of bariatric surgery management in alleviating NAFLD.
Table 3. The effectiveness of bariatric surgery management in alleviating NAFLD.
StudyNumber of PatientsType of SurgeryOutcomes
Weiner et al. (2010) [85]n = 116RYGB, AGB, BPDComplete regression of NAFLD in 82.8%
Moretto et al. (2012) [86]n = 78RYGBImprovement of fibrosis from 44.8% to 30.8%
Tai et al. (2012) [87]n = 21RYGB↓ of steatosis, NASH, and fibrosis
Vargas et al. (2012) [88]n = 26RYGB↓ of steatosis, NASH, and fibrosis
Taitano et al. (2014) [89]n = 160RYGB, AGB75% resolution of steatosis, 90% resolution of NASH, 50% resolution of fibrosis
Lassailly et al. (2015) [90]n = 109RYGB, AGB, BIBResolution of NASH in 85% of patients
Reduction of fibrosis in 34% of patients
Aldoheyan et al. (2017) [91]n = 27RYGB, BPDImprovement of steatosis and fibrosis
Parker et al. (2017) [92]n = 37RYGBRYGB reverses NASH and fibrosis
Esquivel et al. (2018) [93]n = 43SG100% improvement of NAFLD
Schwenger et al. (2018) [84]n = 42RYGB78% resolution of NAFLD
9.5% worsening of fibrosis
Pooler et al. (2019) [94]n = 50RYGBImprovement of steatosis
NAFLD: non-alcoholic fatty liver disease, NASH: non-alcoholic steatohepatitis, RYGB: Roux-en-Y gastric bypass, SG: sleeve gastrectomy, AGB: adjustable gastric banding, BIB: biliointestinal bypass, BPD: biliary-pancreatic diversion, ↓: decrease.
Table 4. Beneficial effects of anti-diabetic drugs on NAFLD.
Table 4. Beneficial effects of anti-diabetic drugs on NAFLD.
StudyNumber of PatientsDurationStudy PopulationDrug NameDoseLiver OutcomesDiabetes Outcomes
Dutour A et al. (2016) [108]n = 4426 weeksObesity, T2DM, NAFLDExenatide5 μg twice daily 4 weeks and after 10 μg/day↓ Hepatic triglyceride↓ Weight
Armstrong MJ et al. (2013) [109]n = 4442Meta-analysisObesity, T2DMLiraglutide1.8 mg/day↓ ALTNot mentioned
Armstrong MJ et al. (2016) [110]n = 712 weeksObesity, NASHLiraglutide1.8 mg/day↓ ALT, AST, DNL↓Weight and adipose mass, ↓ HbA1c and serum levels of glucose, ↓ LDL cholesterol, ↑ insulin sensitivity
Armstrong MJ et al. (2016) [111]n = 2648 weeksObesity, NASHLiraglutide1.8 mg/day↑ NASH resolution↓ Weight, ↓ HbA1c and serum levels of glucose, ↑ HDL cholesterol
Newsome P et al. (2019) [112]n = 957Meta-analysisObesity, with or without T2DMSemaglutideEscalation from 0.05 to 0.4 mg/day↓ ALTNot mentioned
Newsome P et al. (2021) [113]n = 32072 weeksObesity, NASH (F1–F3 fibrosis)Semaglutide0.1, 0.2, or 0.4 mg/day↑ NASH resolution, ↓ ALT, AST↓ Weight, ↓ HbA1c
Cui J et al. (2016) [114]n = 2524 weeksObesity, pre-diabetes, early T2DMSitagliptin100 mg/dayNo changesNo changes
Joy TR et al. (2017) [115]n = 1224 weeksObesity, T2DM, NASHSitagliptin100 mg/dayNo changesNo changes
Neuschwander-Tetri BA et al. (2003) [116]n = 3048 weeksObesity, NASHRosiglitazone4 mg twice daily↓ ALT and AST, ↓ steatosis and inflammatory↓ HbA1c and fasting insulin, ↑ glucose tolerance, ↑ weight
Ratziu V et al. (2008) [117]n = 6352 weeksObesity, NASHRosiglitazone4 mg/day 1 month and after 8 mg/day↓ ALT, ↓ steatosis↓ Fasting glucose and insulin, ↑ weight
Belfort R et al. (2006) [104]n = 5524 weeksObesity, T2DM, NASHPioglitazone45 mg daily↓ ALT and AST, ↓ steatosis and inflammatory↓ Fasting glucose and insulin, ↑ insulin sensitivity, ↑ HDL cholesterol, ↑ weight
Cusi K et al. and Sanyal AJ et al. (2016 + 2010) [118,119]n = 101, n = 24796 weeksObesity, NASHPioglitazone45 mg/day, 30 mg/day↓ ALT and AST, ↓ NAS, ↑ NASH resolution↓ Fasting glucose, insulin and triglyceride, ↑ insulin sensitivity, ↑ HDL cholesterol, ↑ weight
Cusi K et al. (2019) [120]n = 5624 weeksObesity, T2DMCanagliflozin300 mg/dayNo changes↓ Weight, ↓ HbA1c, fasting glucose and insulin, ↑ hepatic insulin sensitivity
Latva-Rasku A et al. (2019) [121]n = 328 weeksObesity, T2DMDapagliflozin10 mg/day↓ Liver lipids, ↓ liver stiffness↓ Weight, ↓ HbA1c, fasting glucose
Shimizu M et al. (2019) [122] n = 5724 weeksT2DM, NAFLDDapagliflozin5 mg/day↓ ALT↓ Weight
Kahl S et al. (2020) [123]n = 4224 weeksObesity, T2DMEmpagliflozin25 mg/day↓ Liver lipids↓ Weight, ↓ glucose
Kuchay MS et al. (2018) [124]n = 2520 weeksT2DM, NAFLDEmpagliflozin10 mg/day↓ ALT, ↓ liver lipidsNo changes
T2DM: type 2 diabetes mellitus, HbA1c: glycated hemoglobin, NAFLD: non-alcoholic fatty liver disease, NASH: non-alcoholic steatohepatitis, NAS: NAFLD Activity Score, AST: aspartate aminotransferase, ALT: alanine aminotransferase, HDL: high-density lipoproteins, LDL: low-density lipoproteins, ↓: decrease, ↑: increase.
Table 5. Ongoing trials of new medications for the treatment of NAFLD.
Table 5. Ongoing trials of new medications for the treatment of NAFLD.
DrugTrial IdentifierNumber of PatientsMechanismT2DM Inclusion
Criteria
Primary Endpoint
ElafibranorNCT027044032000PPARα/δ dual agonistT2DM only with HbA1c ≤ 9%% of patients with NASH resolution without fibrosis worsening at
week 72 from BL, long term liver-related outcomes
SaroglitazarNCT04193982250PPAR-α/γ agonistNAChange in NFS at week 8, 16, and 24
Obeticholic AcidNCT03439254919FXR agonistT2DM only with HbA1c ≤ 9.5%% of patients with improvement of liver fibrosis by ≥1 stage with no worsening of NASH after 18 months
Obeticholic AcidNCT025483512480FXR agonistT2DM only with HbA1c ≤ 9.5%Improvement of liver fibrosis by ≥1stage with no worsening of NASH OR achieving NASH resolution without worsening of liver
fibrosis at month 18 from BL, long term liver-related outcomes
CenicrivirocNCT030287402000CCR2/5 dual antagonistT2DM with HbA1c ≤ 10%Improvement of liver fibrosis by ≥1 stage with no worsening of
NASH after 12 months, long term liver-related outcomes
AramcholNCT041043212000SCD1 inhibitorT2DM with controlled glycemia or prediabetesNASH resolution with no worsening of fibrosis OR fibrosis improvement by ≥1 stage with no worsening of NASH at week 52 from BL, Long term liver-related outcomes
ResmetiromNCT039004292000THR-β agonistT2DM with HbA1c < 9%NASH resolution in patients with F2-F3 fibrosis after 52 weeks, long term liver-related outcomes
BL: baseline, CCR2/5: C-C chemokine receptors type 2 and type 5, FXR: farnesoid X receptor, HbA1c: glycated hemoglobin, LXR: liver X receptor, NA: data not available, NAFLD: non-alcoholic fatty liver disease, NFS: NAFLD fibrosis score, PPAR: peroxisome proliferator-activated receptor, NASH: non-alcoholic steatohepatitis, SCD: stearoyl-CoA desaturase, SGLT: sodium-glucose cotransporter, THR: thyroid hormone receptor, T2DM: type 2 diabetes.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tsamos, G.; Vasdeki, D.; Koufakis, T.; Michou, V.; Makedou, K.; Tzimagiorgis, G. Therapeutic Potentials of Reducing Liver Fat in Non-Alcoholic Fatty Liver Disease: Close Association with Type 2 Diabetes. Metabolites 2023, 13, 517. https://doi.org/10.3390/metabo13040517

AMA Style

Tsamos G, Vasdeki D, Koufakis T, Michou V, Makedou K, Tzimagiorgis G. Therapeutic Potentials of Reducing Liver Fat in Non-Alcoholic Fatty Liver Disease: Close Association with Type 2 Diabetes. Metabolites. 2023; 13(4):517. https://doi.org/10.3390/metabo13040517

Chicago/Turabian Style

Tsamos, Georgios, Dimitra Vasdeki, Theocharis Koufakis, Vassiliki Michou, Kali Makedou, and Georgios Tzimagiorgis. 2023. "Therapeutic Potentials of Reducing Liver Fat in Non-Alcoholic Fatty Liver Disease: Close Association with Type 2 Diabetes" Metabolites 13, no. 4: 517. https://doi.org/10.3390/metabo13040517

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