IMR Press / FBL / Volume 26 / Issue 2 / DOI: 10.2741/4892
Review
Current treatment paradigms and emerging therapies for NAFLD/NASH
Show Less
1 Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
Send correspondence to: Rohit A. Sinha, Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India, Tel- 0522-2494392; E-mail: anthony.rohit@gmail.com
Front. Biosci. (Landmark Ed) 2021, 26(2), 206–237; https://doi.org/10.2741/4892
Published: 1 October 2020
Abstract

Non-alcoholic fatty liver disease (NAFLD) is one the fastest emerging manifestations of the metabolic syndrome worldwide. Non-alcoholic steatohepatitis (NASH), the progressive form of NAFLD, may culminate into cirrhosis and hepatocellular cancer (HCC) and is presently a leading cause of liver transplant. Although a steady progress is seen in understanding of the disease epidemiology, pathogenesis and identifying therapeutic targets, the slowest advancement is seen in the therapeutic field. Currently, there is no FDA approved therapy for this disease and appropriate therapeutic targets are urgently warranted. In this review we discuss the role of lifestyle intervention, pharmacological agents, surgical approaches, and gut microbiome, with regard to therapy for NASH. In particular, we focus the role of insulin sensitizers, thyroid hormone mimetics, antioxidants, cholesterol lowering drugs, incretins and cytokines as therapeutic targets for NASH. We highlight these targets aiming to optimize the future for NASH therapy.

Keywords
NAFLD
NASH
Therapy
Insulin sensitizers
Antioxidants
Statins
Incretins
Cytokines
Bariatric surgery
Gut microbiome
Review
2. INTRODUCTION

Parallel to the global increase in metabolic syndrome, diabetes and obesity, a sharp rise is seen in the prevalence of NAFLD. Non-alcoholic steatohepatitis (NASH) is the clinically serious form of non-alcoholic fatty liver disease (NAFLD), characterized by excessive accumulation of triglycerides (steatosis), inflammation, injury and apoptosis in the liver cells which in extreme cases may lead to cirrhosis and liver cancer (1). NAFLD is currently recognized as the most chronic liver disease, and globally affects around 25% of the adult population (2). It is one the most prominent cause of liver related morbidity and mortality (3).

Owing to its high prevalence and potential risk of progression to cirrhosis and hepatocellular carcinoma (HCC) (4), NASH has become a major health concern worldwide. While it’s adverse hepatic consequences include cirrhosis culminating into liver failure and HCC, it has non-liver associated outcomes such as cardiovascular disease (5), (6). The number of NAFLD cases in the United States is expected to increase from 83.1 million in 2015 to 100.9 million in 2030, a large proportion of which will be NASH (7). This rise will lead to an increase in the number of patients with cirrhosis and end stage liver disease, necessitating liver transplantation (8), (9) and leading to a steep rise in HCC (10).

Despite this high prevalence and growing impact on world health, there are currently no approved treatments for NASH (11). Among all aspects of NASH, the slowest advancement has been seen in the therapeutic field, although a steady progress is seen in understanding disease epidemiology, pathophysiology and identifying therapeutic targets. Even after years of intense research worldwide, presently there is no approved drug for its treatment. Additionally, liver biopsy, a costly and invasive procedure, is still the gold standard for histological evaluation, warranting effective non-invasive imaging and biochemical tests to prevent the need for liver biopsy. This review highlights the treatment strategies for NAFLD/NASH in view of the ongoing rapid progress and unmet challenges, focusing on the critical assessment and understanding of the key elements with regard to therapy for NASH.

3. LIFESTYLE INTERVENTION

Given the fact that presently no drug or surgery is approved for the treatment of NASH, lifestyle modifications (diet, physical activity, and exercise) remain the cornerstone approach for its management. These approaches mainly aim at controlling bodyweight and metabolic disorders (12), (13), (14).

3.1. Physical activity and exercise

Physical activity is recommended for people with NAFLD, as it is a key determinant of metabolic control. People inclined to excessive adiposity, metabolic syndrome and type 2 diabetes (T2D) show higher sedentary behaviour (15). An increase in sedentary time may lead to a predisposition towards NAFLD. Interestingly, the increased breaks to sedentary time are reported to be beneficial for glucose and fatty acid metabolism, and obesity control (16). Several cross-sectional studies have reported lower levels of physical activity in people with NAFLD (17) (18) (19), and that they are more prone to fatigue (20). Individuals with a healthy lifestyle are less predisposed to develop insulin resistance, diabetes and impaired glucose tolerance (21), (22).

Exercise reduces the possibility of developing NASH (23). It lowers the risk of diabetes, hypertension and metabolic syndrome (24), (25). A meta-analysis reports that exercise reduces hepatic fat content, with little or no weight loss (26). It reduces visceral adipose tissue and plasma free fatty acids. The effect of exercise on histological lesions in NASH is not clearly known, however, preliminary data suggest that exercise has an effect on hepatic markers of apoptosis (27). The effect of physical activity shows a dose-effect relationship (24). Vigorous activity is much more beneficial for NASH and fibrosis, as compared to moderate activity (23). The Compendium of Physical Activities defines specific physical activity (PA) based on the rate of energy expenditure. It defines the intensity of physical activity as the ratio of work metabolic rate to a standard resting metabolic rate (MET). One MET (4.184 kJ)·kg1·h1, is defined as the rate of energy expenditure while a person is at rest or the resting metabolic rate during quiet sitting. Activities having MET values between 3 and 5.9 are classified as moderate activity while those with MET values ≥6 are defined as vigorous activities (23).

3.2. Caloric restriction

Excess caloric consumption causes obesity, which is a leading risk factor for NAFLD (2). Caloric restriction (CR) exerts metabolic reprogramming and effective utilization of body fuel, lowering the oxidative damage to cells (28). Carbohydrates (CHO) are the main source of energy in the body. CHO intake is linked to NAFLD (29), and CHO restriction in the diet reduces the glycemic load, and improves insulin resistance and pancreatic β- cell insulin secretion (30). Low CHO diets increase high density lipoprotein (HDL) and reduce serum triglycerides, and glucose (31). In a post hoc analysis of 52 obese and insulin resistant patients, a hypocaloric low-CHO/high-fat diet (40% and 45% total calories per day, respectively) significantly reduced alanine transaminase (ALT) and serum insulin as compared to a hypercaloric high-CHO/low fat diet (60% and 25%, respectively) (32).

The intake of fructose derived from sugars (table sugar) and corn syrup, commonly used in sweetened beverages, has increased (33). Fructose metabolism enhances free radical production, gut permeability, bacterial growth, lipogenesis, and the level of serum lipopolysaccharides (29), (34). A study reported a significant rise in cholesterol (11.4%), serum triglycerides (32.7%), liver fat (150%), visceral adipose fat (25%), and muscle fat (200%) in 47 overweight patients subjected to a daily intake of 1 litre of sugar-sweetened soft drinks up to 6 months (35). High fructose consumption increases the risk of fibrosis in NASH patients (36). A diet rich in omega-3 polyunsaturated fatty acids (PUFA) reduces intrahepatic triglycerides, increases insulin sensitivity (37), and improves steatohepatitis (38), (39). Therefore, CR is recommended for improvement of NAFLD and the overall calorie intake is recommended to be 1,200-1,500 kcal/day for women and 1,500-1,800 kcal/day for men (40).

3.3. Time restricted feeding

Time-restricted feeding (TRF) constitutes a circadian synchronised dietary approach wherein food access is restricted within a particular time window (8-16 hours) (41). A recent study on NAFLD patients demonstrated that TRF helps to significantly achieve weight loss and reduce triglyceride levels after 12 weeks as compared to the control group (42). Furthermore, TRF regimen mice show reduction in severity of hyperinsulinemia, hepatic steatosis and inflammation, despite the consumption of similar quantities of high-fat diet (HFD) as compared to the ad libitum group (43). Interestingly, TRF also mitigates the effect of maternal HFD feeding on fetal hepatic steatosis (44). However, results from clinical trials to test the efficacy of TRF in human NAFLD/NASH are still awaited.

4. PHARMACOLOGICAL THERAPY

Diet and lifestyle intervention measures cannot be successfully or sustainably implemented in most patients. The patients who fail to benefit from lifestyle intervention or those with already advanced disease (significant fibrosis), need pharmacological treatments which are specifically aimed at improving hepatic inflammation, fibrosis and steatohepatitis (45), as illustrated in Figure 1. Steatohepatitis increases liver-related mortality and reduces survival by progression into cirrhosis, liver failure and HCC, therefore, pharmacological therapy is restricted to patients with NASH (46), (45), (47). Several randomized controlled trials (RCTs) have tested pharmacological agents, which show histological improvements in NASH. An effective therapy for NASH is still in infancy; well conducted RCTs aimed at relevant therapeutic targets are needed.

Figure 1

Mechanism of action of lifestyle modification and pharmacological therapy in NAFLD and NASH. NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; TNF: tumor necrosis factor; FXR: farnesoid X receptor; PPAR: peroxisome proliferator-activated receptor.

4.1. Insulin sensitizers

Insulin resistance is a major driving force for excessive fat accumulation in the liver and plays a key role in the initiation and progression of steatohepatitis and fibrosis. Several pharmacological agents listed below are focused to improve insulin resistance.

4.1.1. Thiazolidinediones

Thiazolidinediones (TZD), also known as glitazones, are the strongest evidence-based drugs tested for the treatment of NASH (47). Current guidelines recommend the use of glitazones in NASH (45), (46). Glitazones promote the differentiation of insulin-resistant large pre-adipocytes into small, proliferative, insulin-sensitive adipocytes (49) (50) (51). Upon the induction of lipogenic genes (52) and lipoprotein lipase (LPL), they elevate fatty acid synthesis and uptake in adipose tissue (53), thereby diverting the free fatty acid (FFA) burden towards adipocytes, instead of the liver. Despite the expansion in fat mass, insulin sensitivity is subsequently improved. Alternatively, glitazones upregulate the production of the insulin-sensitizing and anti-steatogenic adipokine, adiponectin (54), which enhances fatty acid beta-oxidation in liver and muscles (55). Thus, they act by redistributing fat from ectopic tissues to the adipose tissue, and by increasing levels of adiponectin. They also contribute to the reduction of insulin resistance by increasing the expression of AMP-activated protein kinase (56), (57) and upregulating glucose transporter type -4 (GLUT-4) in muscle and adipose tissue.

Some studies based on rodents also report anti-inflammatory actions of glitazones, showing decreased hepatic fibrogenesis in response to recurrent liver injury (58), (59). Interestingly, these studies show a reduction in the nuclear expression of peroxisome proliferator-activated receptor (PPAR)-γ, upon activation and trans-differentiation of stellate cells into fibrogenic myofibroblasts (58). PPAR‑γ expression was restored upon treatment with PPAR‑γ agonists and hepatic scar-forming response was elevated (60). This suggests a hepatoprotective effect of glitazones in addition to their insulin-sensitizing property. Rosiglitazone and pioglitazone are the two available glitazones, which exhibit similar cellular action, except lipoprotein metabolism which favours pioglitazone (61). The widespread use of glitazones is restricted by their safety concerns and adverse effects including congestive heart failure (62), bone fractures in women (63), and increased risk of bladder cancer for pioglitazone (64), owing to which, it was withdrawn from the market in France and Germany. As of now, the European Medicines Agency and the FDA continue the licence pioglitazone, except in patients with, or at the risk of developing bladder cancer. Pioglitazone is recommended for treatment of steatohepatitis in biopsy-proven NASH patients. However, the long term benefits and safety issues remain unknown.

4.1.2. Metformin

Another insulin-sensitizing agent is metformin, an oral biguanide, which reduces hepatic glucose production and increases peripheral glucose utilization (65). Metformin is approved for use in patients with type T2D (66). It causes a reduction in endogenous glucose production, activates AMP-activated protein kinase (67) and inhibits the mitochondrial glycerophosphate dehydrogenase shuttle with change in redox state (46). Metformin is considered as a safe drug, however, it is not recommended for the treatment of NASH (69) as there is no proof of its efficacy on hepatic histology.

4.1.3. MSDC-0602K

Pioglitazone, the first-generation insulin sensitizer improved NASH but proved to be of little use owing to its side effects (68). Another second-generation insulin sensitizer, MSDC-0602K, an inhibitor of mitochondrial pyruvate carrier (MPC) with PPAR-γ binding, is reported to enhance lipid oxidation and downregulate de novo lipid synthesis and gluconeogenesis in liver, in vivo and in vitro. MSDC-0602K does not exhibit the side effects associated with the first generation insulin sensitizers (68). MSDC-0602K leads to hepatic histological improvement and reduction in ballooning or lobular inflammation with no increase in fibrosis at 12 months, as reported in the phase 2b, 52-week double-blind study (68).

4.2. Antioxidants

Increased oxidative stress and defected antioxidant defence mechanisms have been studied extensively across the progressive stages of NAFLD, and are believed to contribute to liver injury and development of NASH. Some of the agents with antioxidant activity being used as a therapy for NASH are listed below.

4.2.1. Vitamin E

Vitamin E is a fat soluble compound present in a variety of compounds which belong to two main families, tocopherol and tocotrienol. Vitamin E is present in the phospholipid bilayer of the cell membranes, where it helps to protect against oxidative damage caused by free radicals (55). It protects against mitochondrial toxicity and blocks intrinsic apoptotic pathways, thereby protecting against liver injury (69), (70). It is also reported to have non-antioxidant properties and may alter cell signaling, gene expression (71) or down-regulate nuclear factor kappa B (NF-kB) dependent inflammatory pathways (72).

The largest RCT until now, the PIVENS trial (73) where vitamin E (800 IU/day) was tested along with pioglitazone and placebo in non-diabetic and non-cirrhotic patients with NASH for 2 years, showed significant histological improvement in patients. Vitamin E improved steatosis, inflammation and ballooning, and resolution of NASH was induced in 36% of the patients receiving vitamin E. The safety concerns related to vitamin E include overall mortality (74), haemorrhagic stroke (75), and prostate cancer in males older than 50 years (76). However, the long term safety of vitamin E remains a concern.

4.2.2. Pentoxifylline

Pentoxifylline is a non-selective phosphodiesterase inhibitor used for the treatment of peripheral vascular disease. Animal studies report that pentoxifylline exhibits antioxidant effect (77) and inhibits the production of pro-inflammatory cytokines (78). These effects were confirmed in vivo, where pentoxifylline significantly decreased steatohepatitis in methionine-choline-deficient dietary NASH model (79). In a 1 year long RCT in 55 patients with NASH receiving pentoxifylline or placebo, pentoxifylline improved liver enzyme levels and histological features of NASH, and displayed a marginal effect on fibrosis (80). Good quality human data regarding the use of pentoxifylline in NASH are lacking.

4.3. Lipotoxicity based targets

The progression from steatosis to NASH may be determined by the accumulation of lipotoxic intermediates of triglyceride synthesis and/or lipolysis (81), (82). De novo lipogenesis (DNL) refers to the biochemical process of synthesis of fatty acids from acetyl-CoA subunits commonly produced by carbohydrate catabolism, which occurs primarily in the liver. Fructose is a profound lipogenic substrate which drives DNL, in addition to glucose. Increased hepatic DNL contributes significantly to NASH. The rate-limiting step in DNL is catalysed by acetyl-CoA carboxylase (ACC), which occurs in two major isoforms in mammals, ACC1 and ACC2. The pharmacological inhibition of ACC is another attractive approach to control fatty acid synthesis in lipogenic tissues. GS-0976 is an inhibitor of acetyl-CoA carboxylase. In an open-label study, NASH patients administered with GS-0976 for 12 weeks showed reduction in hepatic DNL, hepatic steatosis and improvement in markers of liver injury (ClinicalTrials.gov no: NCT02856555) (83). NDI-010976, an allosteric inhibitor of ACC1 and ACC2, reduces hepatic de DNL and is reported to improve steatosis, inflammation, and fibrosis in animal models with fatty liver disease (84).

4.3.1. Cholesterol lowering drugs

Cholesterol is a key toxic lipid mediator of liver injury, which may be targeted by specific pharmacological interventions. The synthesis of mono-unsaturated fatty acids such as oleic acid is catalysed by the enzyme stearoyl-CoA desaturase 1 (SCD-1) (85). The deficiency or inhibition of SCD-1 leads to a reduction in liver steatosis and improvement in insulin sensitivity (86), (87), (88). Enhanced SCD-1 activity has been reported in obese patients with NASH (89). A double blind placebo-controlled clinical trial in 60 NAFLD patients reported a reduction in hepatic fat content with the administration of Aramchol, the SCD-1 inhibitor (90).

Statins, also known as HMG-CoA reductase inhibitors, are a class of cholesterol lowering drugs. Statins are reported to show marginal improvement of steatosis (91). They are the first choice agents used to reduce plasma cholesterol and reduce cardiovascular disease (CVD) risk (92). CVD is a leading cause of death in patients suffering from NAFLD and NASH (93). Statins do not show any beneficial effect on liver histology in patients with NASH (91). Therefore, statins are not currently recommended for NASH treatment (46). Concurrent metabolic comorbidities recommend the use of statins for cardiovascular end points in NAFLD patients. A number of studies have focused on the safety of statins in hepatotoxicity (94) as these drugs are reported to lower aminotransferase levels in patients with the metabolic syndrome (95).

4.4. Modulation of nuclear transcription factors

Nuclear transcription factors are also believed to have therapeutic potential for the treatment of NASH. These are molecules, which upon binding to their specific ligand regulate transcription of specific genes.

4.4.1. Farnesoid X receptor (FXR) agonists

The farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily, and has emerged as a key regulator of metabolic pathways including glucose homeostasis and the inflammatory and fibrogenic processes. The hepatic expression level of FXR is inversely correlated with the severity of disease in patients with NASH (96). The FXR is expressed in the liver, kidneys, intestines, and adrenal glands. It is involved in the synthesis and enterohepatic circulation of bile acids and regulates lipoprotein metabolism [97]. FXR activation affects bile acid production, hepatic lipogenesis, cholesterol synthesis and glucose homoeostasis, and protects hepatocytes against bile acid-induced cytotoxicity [98, 99]. This finding is corroborated by FXR-deficient mice fed on a high-fat diet, which exhibit severe hepatic steatosis, necrotic inflammation and fibrosis (100). Furthermore, FXR agonists have been shown to prevent the development of NASH in rodent models of diet-induced NASH, where they also promoted the resolution of steatohepatitis and fibrosis (101). Therefore, a number of synthetic FXR agonists are being tested for the treatment of NASH and other hepatic disorders (102), (103).

4.4.1.1. Obeticholic acid

Obeticholic acid (OCA) is a modified bile acid which is derived from the primary human bile acid, chenodeoxycholic acid. It stimulates the FXR in humans (104). The FXR plays a key role in the synthesis and entero-hepatic circulation of bile acids (105) and is expressed in the liver, adrenal glands, kidneys and intestines. OCA is a synthetic 6α-ethyl derivative of chenodeoxycholic acid, the primary human bile acid. It is a potent agonist of the FXR, a nuclear hormone receptor which regulates glucose and lipid metabolism. This chemical modification stimulates the activity of FXR approximately 100-fold more than the natural FXR agonist in humans (106), chenodeoxycholic acid. It shows high selectivity and minimal activity with other bile acid receptors (107).

The FLINT trial, a phase 2b study (108) and double-blind, placebo-controlled, randomized clinical trial in the USA, assessed NASH patients without cirrhosis and a NAFLD activity score (NAS) ≥4, for 72 weeks. Improvement in centrally scored liver histology by 2 points in NASH was observed, without any worsening of fibrosis. In FLINT trial, the administration of OCA was associated with significantly great increase in the concentration of total cholesterol and low-density lipoprotein cholesterol (LDLc) and decrease in the concentration of high-density lipoprotein cholesterol (HDLc) as compared to placebo. However, the increase in LDLc concentrations observed upon treatment with OCA were reversed with concomitant statin therapy (e.g. atorvastatin).

Another ongoing phase 3 REGENERATE trial (109) aims to compare the effects of OCA and placebo in the histological improvements and clinical outcomes in NASH and liver fibrosis stage 2 and 3. The patients are randomized and treated with OCA 10 mg, OCA 25 mg and placebo. The primary analysis included 931 patients suffering from F2-F3 fibrosis (311 placed in the placebo group, 312 in OCA 10 mg treated group and 308 in OCA 25 mg treated group). An improvement in fibrosis was seen in 37 (12%) patients of the placebo group, 55 (18%) patients of the OCA 10 mg group (P = 0.045), and 71 (23%) patients of the OCA 25 mg group (P = 0.0002). The resolution of NASH was not attained in any of the patients.

4.4.1.2. Tropifexor (LJN-452)

Tropifexor (LJN-452) is a very potent non-bile acid agonist of FXR, which is reported to induce target genes at very low concentrations. Tropifexor has been shown to be very effective in preclinical NASH models (110). An ongoing phase 2 RCT, FLIGHTFXR, is evaluating the efficacy and safety of tropifexor and cenicriviroc in patients with fibrosis and biopsy-proven NASH (111).

4.4.2. Peroxisome proliferator-activated receptors (PPARs)

PPARs are a group of nuclear receptors, which are expressed in the liver, adipose tissue, heart, skeletal muscle and kidney, and transcriptionally regulate multiple metabolic processes including β-oxidation, lipid transport and gluconeogenesis (112). They are classified into three isotypes, designated as PPARα, PPARβ (also known as PPARδ) and PPARγ, which differ in tissue distribution; however, they target the same segment of DNA. The PPARs are known to heterodimerize with the retinoid X receptor (RXR) to regulate target gene transcription.

In the liver, PPARα upregulates several enzymes involved in mitochondrial and peroxisomal fatty acid oxidation, microsomal ω‑oxidation, and ketogenesis (113), (114), and therefore shifts the hepatic metabolism towards lipid oxidation. The activation of PPARα upregulates the expression of lipoprotein lipase (LPL) while downregulating the hepatic secretion of APOCIII, a LPL inhibitor (115), thereby elevating plasma triglyceride clearance. In vitro and in vivo studies report that PPARα suppresses the secretion of interleukin 1 (IL‑1), IL‑6 and tumour necrosis factor (TNF), and intercellular adhesion molecule 1 (ICAM1) and vascular cell adhesion molecule 1 (VCAM1) independent of direct DNA binding (116), (117). These effects were peroxisome proliferator response elements (PPRE)-independent, yet they were hepatoprotective against methionine–choline-deficient diet-induced inflammation and fibrosis, without altering fatty acid oxidation and lipid accumulation (118).

4.4.2.1. Elafibranor

Elafibranor is a PPAR-alpha/delta (α/δ) agonist, known to regulate lipid and insulin metabolism in NAFLD and NASH (119). A phase 2b, double blind RCT, the GOLDEN study (Europe and USA), compared 80 mg and 120 mg elafibranor daily to placebo in 276 patients with biopsy-proven, non-cirrhotic NASH with NAS ≥3 and ≥1 point, for 52 weeks. NASH was resolved, without the fibrosis worsening in the higher proportion of patients of the 120 mg elafibranor group as compared to the placebo group (19% vs 12%; odds ratio = 2.31; P = .045). However, reversal of NASH was not achieved (119).

An ongoing phase 3 study, RESOLVE-IT (https://clinicaltrials.gov/ct2/show/NCT0270443) includes 2,000 patients with NASH having NAS ≥4, with ≥1 of each component of score, and F1-F3 fibrosis. The trial aims at the histological improvement in NASH, evident by the resolution of NASH without any worsening of fibrosis at 72 weeks, and to evaluate cirrhosis, liver related clinical outcomes, and mortality at 4 years. The results of this trial are due in December 2021.

4.4.2.2. Saroglitazar

Another dual-PPAR agonist, saroglitazar, has been shown to improve lipid and glucose parameters by predominant PPAR-α and moderate PPAR-γ agonist activity. A phase-2 RCT determined the efficacy of saroglitazar magnesium as compared to placebo in patients with NASH (120). A significant reduction was observed in mean ALT levels from baseline to week 16, with 1 mg (−27.3%), 2 mg (−33.1%) and 4 mg (−44.3%) saroglitazar as compared to placebo (4.1%) (P < 0.001 for all groups). A high proportion of patients showed ≥50% reduction in the mean ALT levels from the baseline to week 16 with 4 mg saroglitazar as compared to the placebo (51.8% versus 3.5%; P < 0.0001). At week 16, a significant proportion of patients showed >30% reduction in the liver fat content with 4 mg saroglitazar as compared to placebo (40.7% vs 8%, P = 0.006).

4.4.3. Thyroid hormone receptor (THR)

Thyroid hormone receptor β (THR-β) is known to be highly expressed in the hepatic cells. It plays a key role in regulation of the metabolic pathways which are known to be damaged in NASH (121). Animal studies have reported a crucial role of THR-β in reducing triglycerides and cholesterol, improving insulin sensitivity, promoting liver regeneration, and reducing apoptosis. Evidences indicate that NASH may be, in part, caused by hepatic hypothyroidism, which is higher in patients with NASH (121), (122).

4.4.3.1. Resmetirom (MGL-3196)

Encouraged from the earlier preclinical studies Thyroid hormone mimetics are being actively pursued as possible anti-NASH therapy (123). Resmetirom (MGL-3196) is an orally active agonist of THR. It is liver-directed and about 28 times more selective for THR-β versus THR-α than triiodothyronine (124). In NASH, this selectivity for THR-β is believed to provide metabolic benefits of thyroid hormone which are mediated by the liver, while avoiding the unwanted effects of excess thyroid hormone in the heart and bone which are mainly mediated through THR-α (121). In preclinical animal models of NASH, resmetirom and some other thyroid analogues were shown to reduce hepatic triglycerides, hepatic steatosis, lipid peroxidation, inflammatory and fibrosis markers, along with ALT levels (124), (121). A multicentre, randomised, double-blind, placebo-controlled, phase clinical 2 trial based on Resmetirom treatment (ClinicalTrials.gov Identifier: NCT03900429) resulted in the significant reduction of hepatic fat in patients with NASH, after 12 weeks and 36 weeks of treatment. The adverse effects of treatment in study groups were mild or moderate, except high occurrence of transient mild diarrhoea and nausea observed in the group treated with Resmetirom (125).

VK2809 is another tissue and receptor subtype selective agonist of THR-β which is specific to the liver tissue and hold promising therapeutic potential. It belongs to the pro-drug family which are cleaved in vivo and release potent thyromimetics. It is believed to improve cholesterol and lipoprotein levels through multiple mechanisms including induction of gene expression of genes associated with lipid metabolism. A phase 2, randomized, double-blind, placebo-controlled, multicenter study has been designed to assess the efficacy, safety, and tolerability of VK2809 in lowering LDL-C and liver fat content when administered in patients with primary hypercholesterolemia and NAFLD for 12 weeks, followed by a 4-week off-drug phase (ClinicalTrials.gov Identifier: NCT02927184). Patients receiving 5mg daily doses of VK2809 demonstrated a statistically significant median reduction of 53.8% in liver fat. Of the patients receiving VK2809, 88% showed hepatic fat reduction ≥30% at 12 weeks. At all doses studied, VK2809 was safe and well tolerated and no adverse effects were reported among patients (126).

4.5 Gastrointestinal hormones (Incretins)

Evidences indicate that peptide hormones from the Langerhans cells of distal small intestine serve as incretins, which trigger the release of insulin by the pancreatic beta cells. These gut derived incretin hormones are potential targets for the treatment of NASH.

4.5.1. Glucagon-like peptide (GLP)-1 agonist

Glucagon-like peptide (GLP)-1 is an intestinal hormone released from the foregut in response to food consumption. GLP-1 exhibits a glucose lowering action by its glucose-dependent ability to stimulate insulin secretion and inhibit glucagon secretion from the pancreas. It also delays gastric emptying and suppresses appetite. These metabolic effects of GLP-1 contribute to its anti-NASH activity (127). Its endogenous form is, however, rapidly degraded in vivo by the enzyme dipeptidyl peptidase-4 (DPP-4). GLP-1 secretion is reported to be impaired in patients with NAFLD and NASH, suggesting potential role of GLP-1 agonists in NAFLD treatment (128). The GLP-1 agonists mimic gastrointestinal endogenous GLP-1 hormone and have increased resistance to DPP-4 (129). They have a beneficial effect on insulin resistance and weight control (130). The presence of GLP-1 receptor in hepatocytes has been reported by several studies; therefore GLP-1 agonists may exert their direct effect on liver. Studies suggest that GLP-1 agonists reduce fat accumulation by activating macroautophagy and chaperone mediated autophagy (131).

A pilot study reports Liraglutide, a GLP-1 agonist, to improve NASH histology upon daily injection (132). Semaglutide is another GLP-1 agonist under study (NCT02970942), having the advantage that it requires weekly dosing only. Therapies for NASH, aimed at using a combination of GLP-1 with glucagon or GLP-2 are in early development stages.

4.6. Cytokines

Cytokines are the cell signaling molecules produced by a variety of cells in the body, including liver cells (133). They are crucial mediators of inflammation related disorders. They include several subfamilies such as including interferons, interleukins (IL), transforming growth factors (TGF), TNF, colony-stimulating factors, and chemokines. Lipid accumulation and inflammation are crucial in NAFLD; therefore, cytokines may play a key role in the pathogenesis of NAFLD by stimulating hepatic inflammation, steatosis, cell apoptosis and necrosis, and by inducing fibrosis. The pro-inflammatory cytokines, IL family and TNFα are reported to control the key features of liver disease.

4.6.1. Tumor necrosis factor-alpha (TNF-α)

TNF-α is a pro-inflammatory cytokine secreted by monocytes/macrophages, neutrophils, and T-cells, and other tissues, such as the adipose tissue, neuronal tissue and endothelium. It is associated with obesity and related insulin resistance (134). Dietary and genetic (ob/ob) mice models of obesity lacking TNFα showed improved insulin sensitivity (135). Clinical investigations show that TNFα expression is increased in obesity and decreases on weight loss (136), (137) indicating its role in obesity and metabolic dysregulation (138). A Spanish study based on 52 obese patients showed that liver disease severity correlated with TNF-α gene expression in adipose and liver tissue (139) Overexpression of TNF-α mRNA was observed in the adipose tissue and liver of NASH patients. An Italian study reported increased prevalence of TNF-α polymorphism in NAFLD patients as compared to the controls (140). Hence, TNFα is proposed as a potential therapeutic target to prevent the consequences of metabolic syndrome.

The anti-TNFα drug, thalidomide helped to improve the hepatic alterations caused by a high fat diet in mice (141). Anti-TNFα antibodies decreased inflammation, necrosis, and fibrosis in an experimental rat model of NASH (142). The neutralization of TNFα by Infliximab showed substantial reduction in steatosis, in ob/ob mice (143). Although animal models of NAFLD show encouraging therapeutic potential of TNF-α inhibition, its role in humans is controversial. Hotamisligil et al (144) reported a relationship between the expression of TNF-α and insulin resistance in NASH. A positive correlation has been reported between circulating TNF-α levels and the degree of liver fibrosis, in patients with NASH (144). Increased expression of TNF-α has been observed in the liver and adipose tissue of NASH patients showing significant fibrosis as compared to those with slight or non-existent fibrosis (145). Hui et al (146) showed increased TNFα levels in steatohepatitis subjects as compared to the controls. However, some studies showed no correlation between insulin resistance and TNFα levels (147), (148).

4.6.2. Transforming growth factor-beta (TGF-β)

Transforming growth factor-beta (TGF-β) is an immunosuppressive and pro-fibrotic cytokine (149). TGF-β1 mediates the activation of stellate cells and plays a key role in hepatic fibrosis (150) (151) (152). Its expression is upregulated in experimental hepatic fibrosis models (153), (154), (155). Furthermore, the TGF-β1 mRNA expression is increased in patients with liver fibrosis (156), (157), (158). Stärkel et al (159) reported that upregulation of TGF-β1 is an early step in progressive fibrotic steatohepatitis. Hasegawa and co-workers showed that the levels of TGF-β1 are increased in NASH patients. Serum TGF-β1 levels may help to distinguish NASH patients from the spectrum of NAFLD (160). Liver fibrosis is believed to be a strong predictor of mortality in patients with NAFLD, therefore, there is growing interest in developing therapeutics to target elements in fibrogenesis. TGF-β is a key pro-fibrogenic cytokine, which may be a promising target to treat fibrosis. High levels of TGF-β, caused by chronic liver damage, result in activation of stellate cells to myofibroblasts and massive hepatocyte cell death contributing to the promotion of liver fibrosis. TGF-β signaling induces Smad-dependent and TAK1-dependent signaling which regulates cell survival, proliferation, fibrosis, and tumorigenesis [161]. Galunisertib (LY2157299) is a promising anti-fibrotic TGF-β inhibitor which inhibits SMAD2 phosphorylation and blocks collagens deposition [162]. Although several approaches to inhibit TGF-β signalling in animal models have yielded promising results, the complex role of TGF- β in the liver in cell proliferation, carcinogenesis and immune modulation is a major problem [163, 164] to developing anti- TGF-β based therapy for NASH.

Cenicriviroc (CVC) is a dual antagonist of chemokine receptor type 2 (CCR2) and type 5 (CCR5), located on hepatic stellate cells and Kupffer cells is being targeted for anti-fibrosis therapy [165]. It blocks overactive inflammatory signalling and disrupts signalling that activates stellate cells thereby targeting both inflammation and fibrogenesis. CVC is orally administered in daily tablets (150 mg) and has a plasma life of 30-40 hours. CENTAUR, a phase 2b, 24-month study included 189 patients with biopsy-proven NASH, with an NAS ≥4 and stage 1-3 fibrosis. The patients were randomized 2:1:1 in three arms as follows: A) continuous administration of 150-mg CVC for 24 months, B) administration of placebo for 12 months followed by an additional 12 months of CVC, and C) administration of placebo for 24 months. The expected primary outcome of ≥2 point decrease in NAFLD activity score without any worsening in fibrosis at 1 year was not achieved. However, the improvement in liver fibrosis without worsening of NASH was achieved in 20% from the treatment group, along with lower levels of interleukin-6, C-reactive protein and fibrogen [166]. Currently, another randomized, double-blind, placebo-controlled, multi-centre phase 3 study, AURORA (https://clinicaltrials.gov/ct2/show/NCT03028740) is evaluating the safety and efficacy of CVC in treatment of fibrosis, in NASH.

4.6.3. Interleukin-11 (IL-11)

Hepatic stellate cells (HSCs) give rise to upto 95% of liver myofibroblasts (167), and are crucial to fibrosis, inflammation and parenchymal dysfunction in NASH (168) (169). They are a major source of proinflammatory myofibroblasts, and therefore, their inhibition or reversal of transformation in the liver, may be a potential therapy for treatment of NASH. Interleukin-11 (IL11) was recently identified as a key factor for pulmonary and cardiovascular fibroblast-to-myofibroblast transformation (170, 171). ERK-dependent IL11 signaling is critical for HSC transformation (154), (155), (172).

Recently, a study by Widjaja et al based on hepatocytes, HSCs, and mouse models, explored the role of IL-11 signaling in NASH pathogenesis (172). They reported that HSCs express high levels of interleukin 11 subunit alpha (IL11RA) and showed a proinflammatory role of IL11 in the liver. The ALT levels reversed from approximately 700 U/L to normal within 3 weeks, upon pharmacologic targeting of IL11 signaling (172). They showed that neutralizing antibodies which block IL11 signaling, caused reduction in fibrosis, steatosis, hepatocyte apoptosis and inflammation, in mice with diet-induced liver steatosis and fibrosis. They also reported that hepatocyte damage is prevented, or reversed, by genetic or antibody-mediated inhibition of IL11. Therefore, targeting IL11 to reverse liver fibrosis may be a beneficial therapy for NASH.

5. BARIATRIC SURGERY

Bariatric surgery or a weight loss surgery may be opted to achieve weight loss in NAFLD/NASH patients. Weight loss through diet and exercise may be difficult to achieve or sustain. Bariatric surgery helps to achieve long-term weight loss. It also helps to improve the metabolic functioning of lipid metabolism and inflammatory pathways associated with pathophysiology of NAFLD (173), (174). Bariatric surgery is currently recommended in patients with BMI ≥40 kg/m2 having no comorbidities, or with a BMI 35 to 39.9 kg/m2 with any serious comorbidity such as T2D, hypertension, NAFLD and NASH (175), (176). A review based on 29 studies showed significant improvement in ALT, aspartate aminotransferase (AST), Gamma-glutamyl transferase (GGT) and histology in patients undergoing bariatric surgery (177). A meta-analysis reported improvement in steatosis, steatohepatitis, and fibrosis following weight loss after bariatric surgery (178). Bariatric surgery is an invasive procedure and is therefore associated with risks such as adjustable gastric banding (AGB), and dietary complications including nutritional deficiencies, postprandial hyperinsulinemic hypoglycaemia, etc. (179), (180), (181), (182), (183). A review by le Roux and Heneghan has outlined the long term complications associated with bariatric surgery (179).

6. GUT MICROBIOME

The gut microbiome produces substances which help to regulate immunity, nutrition, homeostasis, and several metabolic pathways (184), (185), (186). The gut microbiota interact with liver through the ‘liver-gut axis’, involving specific metabolites such as bile acids (BAs), short-chain fatty acids (SCFAs), and lipopolysaccharides (LPS) (187). Several bacteria are known to be associated with lipid and carbohydrate metabolism. Evidences indicate the effect of gut microbiota on liver function, which contribute to energy metabolism, obesity, T2DM and NAFLD (188), (189). Therefore, gut microbiota may regulate disorders linked to energy metabolism (184), (190), (191) Gut microbiota dysbiosis may be caused by the changes in the environment of the host such as diet, alcohol intake, genetic factors, and antibiotics (192). Gut microbiota dysbiosis plays a pivotal role in the pathogenesis of metabolic disorders including NAFLD (193)

Studies based on animal models suggest that probiotics, prebiotics and synbiotics have a modulatory effect on gut microbiota. The oral administration of probiotics is shown to improve abnormal lipid metabolism and dysbiosis of gut microbiota (194). Supplementation of prebiotics and synbiotics improved hypercholesterolemia-associated hepatic changes in rats by regulating genes involved in β-oxidation and lipogenesis, including PPAR-α, carnitine palmitoyltransferase 1 (CPT-1), sterol regulatory element-binding protein 1c (SREBP-1c), fatty acid synthase (FAS) and malic enzyme (ME) (195). Improvement in hepatic inflammation and insulin resistance was observed in rats treated with synbiotics. Also, a reduction in the amount of Gram-negative Enterobacteriales and Escherichia coli was seen in the colonic mucosa.

Fecal microbiota transplantation (FMT) is a method for transplantation of fecal bacteria from a healthy donor to re-populate gut microbiota in patients for the treatment of several diseases (196), (197), (198). FMT is being widely explored for the treatment of NAFLD/NASH. Intrahepatic lipid accumulation, insulin resistance and pro-inflammatory cytokines were improved in mouse models, by FMT (199), (200). In a RCT based on patients with metabolic syndrome, increase in insulin sensitivity was observed after 6 weeks of receiving gut microbiota from healthy Caucasian males (201). FMT may also be developed as a potential therapeutic method for NASH.

7. SUMMARY

While there is no FDA-approved medication for NAFLD/NASH, dietary and lifestyle intervention is the mainstay of treatment. Several medications are in pipeline of therapy of NASH, holding promise for a successful therapy in future. As of now, the first line drugs such as pioglitazone and vitamin E remain the strategy for disease management in patients.

8. ACKNOWLEDGMENTS

This work was supported by the ICMR (59/05/2019/ONLINE/BMS/TRM), SERB (SRG/2019/000398), and Wellcome Trust/DBT India Alliance Fellowship (IA/I/16/2/502691) awarded to Sinha RA.

Abbreviations
Abbreviation Expansion
NASH

non-alcoholic steatohepatitis

NAFLD

non-alcoholic fatty liver disease

HCC

hepatocellular carcinoma

CR

caloric restriction

CHO

carbohydrates

HDL

high density lipoprotein

ALT

alanine transaminase

PUFA

polyunsaturated fatty acids

TRF

time-restricted feeding, HFD: high-fat diet

RCT

randomized controlled trial

TZD

thiazolidinediones

LPL

lipoprotein lipase

FFA

free fatty acid

PPAR

peroxisome proliferator-activated receptor

GLUT-4

glucose transporter type-4

T2D

type 2 diabetes

MPC

mitochondrial pyruvate carrier

NF-kB

nuclear factor kappa B

SCD

stearoyl-CoA desaturase, CVD: cardiovascular disease

FXR

farnesoid X receptor

OCA

obeticholic acid

TNF

tumour necrosis factor

VCAM1

vascular cell adhesion molecule 1

IL-1

interleukin 1

ICAM1

intercellular adhesion molecule 1

THR

thyroid hormone receptor

GLP-1

glucagon-like peptide

DPP-4

dipeptidyl peptidase-4

TGF

transforming growth factors

HSCs

Hepatic stellate cells

IL-11

Interleukin-11

AGB

adjustable gastric banding

Bas

bile acids

SCFAs

short-chain fatty acids

CPT-1

carnitine palmitoyltransferase 1

SREBP-1c

sterol regulatory element-binding protein 1c

FAS

fatty acid synthase

ME

malic enzyme

FMT

Fecal microbiota transplantation

References
[1]
MarginiC DufourJ. F The story of HCC in NAFLD: from epidemiology, across pathogenesis, to prevention and treatmentLiver Int 201636331724DOI: 10.1111/liv.13031
[2]
YounossiZ. M KoenigA. B AbdelatifD FazelY HenryL WymerM Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomesHepatology20166417384DOI: 10.1002/hep.28431
[3]
LoombaR SanyalA. J The global NAFLD epidemicNat Rev Gastroenterol Hepatol2013101168690DOI: 10.1038/nrgastro.2013.171
[4]
RazaS RajakS AnjumB SinhaR. A Molecular links between non-alcoholic fatty liver disease and hepatocellular carcinomaHepatoma Res2019542DOI: 10.20517/2394-5079.2019.014
[5]
LindenmeyerC. C McCulloughA. J The Natural History of Nonalcoholic Fatty Liver Disease-An Evolving ViewClin Liver Dis20182211121DOI: 10.1016/j.cld.2017.08.003
[6]
RinellaM. E SanyalA. J Management of NAFLD: a stage-based approachNat Rev Gastroenterol Hepatol2016134196205DOI: 10.1038/nrgastro.2016.3
[7]
EstesC RazaviH LoombaR YounossiZ SanyalA. J Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of diseaseHepatology2018671123133DOI: 10.1002/hep.29466
[8]
GoldbergD DitahI. C SaeianK LalehzariM AronsohnA GorospeE. C CharltonM Changes in the Prevalence of Hepatitis C Virus Infection, Nonalcoholic Steatohepatitis, and Alcoholic Liver Disease Among Patients With Cirrhosis or Liver Failure on the Waitlist for Liver TransplantationGastroenterology2017152510901099 e1DOI: 10.1053/j.gastro.2017.01.003
[9]
WongR. J AguilarM CheungR PerumpailR. B HarrisonS. A YounossiZ. M AhmedA Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United StatesGastroenterology2015148354755DOI: 10.1053/j.gastro.2014.11.039
[10]
MittalS El-SeragH. B SadaY. H KanwalF DuanZ TempleS MayS. B KramerJ. R RichardsonP. A DavilaJ. A Hepatocellular Carcinoma in the Absence of Cirrhosis in United States Veterans is Associated With Nonalcoholic Fatty Liver DiseaseClin Gastroenterol Hepatol201614112431 e1DOI: 10.1016/j.cgh.2015.07.019
[11]
MuthiahM. D SanyalA. J Current management of non-alcoholic steatohepatitisLiver Int202040Suppl 18995DOI: 10.1111/liv.14355
[12]
TeedeH DeeksA MoranL Polycystic ovary syndrome: a complex condition with psychological, reproductive and metabolic manifestations that impacts on health across the lifespanBMC Med2010841DOI: 10.1186/1741-7015-8-41
[13]
PreissD SattarN Non-alcoholic fatty liver disease: an overview of prevalence, diagnosis, pathogenesis and treatment considerationsClin Sci (Lond)2008115514150DOI: 10.1042/CS20070402
[14]
MahadyS. E GeorgeJ Exercise and diet in the management of nonalcoholic fatty liver diseaseMetabolism2016658117282DOI: 10.1016/j.metabol.2015.10.032
[15]
DunstanD. W SalmonJ HealyG. N ShawJ. E JolleyD ZimmetP. Z OwenN SteeringC. AusDiab Association of television viewing with fasting and 2-h postchallenge plasma glucose levels in adults without diagnosed diabetesDiabetes Care200730351622DOI: 10.2337/dc06-1996
[16]
HealyG. N DunstanD. W SalmonJ CerinE ShawJ. E ZimmetP. Z OwenN Breaks in sedentary time: beneficial associations with metabolic riskDiabetes Care20083146616DOI: 10.2337/dc07-2046
[17]
PerseghinG LattuadaG CobelliF. De RagognaF NtaliG EspositoA BelloniE CanuT TerruzziI ScifoP MaschioA. Del LuziL Habitual physical activity is associated with intrahepatic fat content in humansDiabetes Care20073036838DOI: 10.2337/dc06-2032
[18]
GeorgeA. St BaumanA JohnstonA FarrellG CheyT GeorgeJ Independent effects of physical activity in patients with nonalcoholic fatty liver diseaseHepatology20095016876DOI: 10.1002/hep.22940
[19]
Zelber-SagiS Nitzan-KaluskiD GoldsmithR WebbM ZvibelI GoldinerI BlendisL HalpernZ OrenR Role of leisure-time physical activity in nonalcoholic fatty liver disease: a population-based studyHepatology200848617918DOI: 10.1002/hep.22525
[20]
NewtonJ. L JonesD. E HendersonE KaneL WiltonK BurtA. D DayC. P Fatigue in non-alcoholic fatty liver disease (NAFLD) is significant and associates with inactivity and excessive daytime sleepiness but not with liver disease severity or insulin resistanceGut200857680713DOI: 10.1136/gut.2007.139303
[21]
SnowlingN. J HopkinsW. G Effects of different modes of exercise training on glucose control and risk factors for complications in type 2 diabetic patients: a meta-analysisDiabetes Care20062911251827DOI: 10.2337/dc06-1317
[22]
ThomasD. E ElliottE. J NaughtonG. A Exercise for type 2 diabetes mellitusCochrane Database Syst Rev(3)2006CD002968DOI: 10.1002/14651858. CD002968.pub2
[23]
KistlerK. D BruntE. M ClarkJ. M DiehlA. M SallisJ. F SchwimmerJ. B GroupN. C. R Physical activity recommendations, exercise intensity, and histological severity of nonalcoholic fatty liver diseaseAm J Gastroenterol20111063460-8quiz 469DOI: 10.1038/ajg.2010.488
[24]
WenC. P WaiJ. P TsaiM. K YangY. C ChengT. Y LeeM. C ChanH. T TsaoC. K TsaiS. P WuX Minimum amount of physical activity for reduced mortality and extended life expectancy: a prospective cohort studyLancet20113789798124453DOI: 10.1016/S0140-6736(11)60749-6
[25]
LeeI. M ShiromaE. J LobeloF PuskaP BlairS. N KatzmarzykP. T WorkingG. Lancet Physical Activity Series Effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancyLancet2012380983821929DOI: 10.1016/S0140-6736(12)61031-9
[26]
KeatingS. E HackettD. A GeorgeJ JohnsonN. A Exercise and non-alcoholic fatty liver disease: a systematic review and meta-analysisJ Hepatol201257115766DOI: 10.1016/j.jhep.2012.02.023
[27]
FealyC. E HausJ. M SolomonT. P PagadalaM FlaskC. A McCulloughA. J KirwanJ. P Short-term exercise reduces markers of hepatocyte apoptosis in nonalcoholic fatty liver diseaseJ Appl Physiol (1985)2012113116DOI: 10.1152/japplphysiol.00127.2012
[28]
AndersonR. M WeindruchR Metabolic reprogramming, caloric restriction and agingTrends Endocrinol Metab2010213134141DOI: 10.1016/j.tem.2009.11.005
[29]
JensenT AbdelmalekM. F. SullivanS NadeauK. J GreenM RoncalC NakagawaT KuwabaraM SatoY KangD. H TolanD. R Sanchez-LozadaL. G RosenH. R LanaspaM. A DiehlA. M JohnsonR. J Fructose and sugar: A major mediator of non-alcoholic fatty liver diseaseJ. Hepatol201868510631075DOI: 10.1016/j.jhep.2018.01.019
[30]
LudwigD. S EbbelingC. B The Carbohydrate-Insulin Model of Obesity: Beyond "Calories In, Calories Out"JAMA Intern Med2018178810981103DOI: 10.1001/jamainternmed.2018.2933
[31]
DongT GuoM ZhangP SunG ChenB The effects of low-carbohydrate diets on cardiovascular risk factors: A meta-analysisPLoS ONE2020151e0225348DOI: 10.1371/ journal.pone.0225348
[32]
RyanM. C AbbasiF LamendolaC CarterS McLaughlinT. L Serum alanine aminotransferase levels decrease further with carbohydrate than fat restriction in insulin-resistant adultsDiabetes Care2007305107580DOI: 10.2337/dc06-2169
[33]
TappyL LeK. A Metabolic effects of fructose and the worldwide increase in obesityPhysiol Rev20109012346DOI: 10.1152/physrev.00019.2009
[34]
VosM. B LavineJ. E Dietary fructose in nonalcoholic fatty liver diseaseHepatology2013576252531DOI: 10.1002/hep.26299
[35]
MaerskM BelzaA Stodkilde-JorgensenH RinggaardS ChabanovaE ThomsenH PedersenS. B AstrupA RichelsenB Sucrose-sweetened beverages increase fat storage in the liver, muscle, and visceral fat depot: a 6-mo randomized intervention studyAm J Clin Nutr20129522839DOI: 10.3945/ajcn.111.022533
[36]
AbdelmalekM. F SuzukiA GuyC Unalp-AridaA ColvinR JohnsonR. J DiehlA. M ResearchN. Nonalcoholic Steatohepatitis Clinical Increased fructose consumption is associated with fibrosis severity in patients with nonalcoholic fatty liver diseaseHepatology2010516196171DOI: 10.1002/hep.23535
[37]
StorlienL. H KraegenE. W ChisholmD. J FordG. L BruceD. G PascoeW. S Fish oil prevents insulin resistance induced by high-fat feeding in ratsScience198723748178858DOI: 10.1126/science.3303333
[38]
SekiyaM YahagiN MatsuzakaT NajimaY NakakukiM NagaiR IshibashiS OsugaJ YamadaN ShimanoH Polyunsaturated fatty acids ameliorate hepatic steatosis in obese mice by SREBP-1 suppressionHepatology2003386152939DOI: 10.1016/j.hep.2003.09.028
[39]
LevyJ. R CloreJ. N StevensW Dietary n-3 polyunsaturated fatty acids decrease hepatic triglycerides in Fischer 344 ratsHepatology200439360816DOI: 10.1002/hep.20093
[40]
MarchesiniG PettaS GraveR. Dalle Diet, weight loss, and liver health in nonalcoholic fatty liver disease: Pathophysiology, evidence, and practice. Hepatology,20166320322043DOI: 10.1002/hep.28392
[41]
PattersonR. E LaughlinG. A LaCroixA. Z HartmanS. J NatarajanL SengerC. M MartinezM. E VillasenorA SearsD. D MarinacC. R GalloL. C Intermittent Fasting and Human Metabolic Health.J Acad Nutr Diet20151158120312DOI: 10.1016/j.jand.2015.02.018
[42]
CaiH QinY. L ShiZ. Y ChenJ. H ZengM. J ZhouW ChenR. Q ChenZ. Y Effects of alternate-day fasting on body weight and dyslipidaemia in patients with non-alcoholic fatty liver disease: a randomised controlled trialBMC Gastroenterol2019191219DOI: 10.1186/s12876-019-1132-8
[43]
ChungH ChouW SearsD. D PattersonR. E WebsterN. J ElliesL. G Time-restricted feeding improves insulin resistance and hepatic steatosis in a mouse model of postmenopausal obesityMetabolism2016651217431754DOI: 10.1016/j.metabol.2016.09.006
[44]
UpadhyayA AnjumB GodboleN. M RajakS ShuklaP TiwariS SinhaR. A GodboleM. M Time-restricted feeding reduces high-fat diet associated placental inflammation and limits adverse effects on fetal organ developmentBiochem Biophys Res Commun20195142415421DOI: 10.1016/j.bbrc.2019.04.154
[45]
RatziuV BellentaniS Cortez-PintoH DayC MarchesiniG A position statement on NAFLD/NASH based on the EASL 2009 special conferenceJ Hepatol201053237284DOI: 10.1016/j.jhep.2010.04.008
[46]
ChalasaniN YounossiZ LavineJ. E DiehlA. M BruntE. M CusiK CharltonM SanyalA. J GastroenterologicalA. American LiverD. American Association for the Study of ofG. American College The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Gastroenterological Association, American Association for the Study of Liver Diseases, and American College of GastroenterologyGastroenterology201214271592609DOI: 10.1053/j.gastro.2012.04.001
[47]
VuppalanchiR ChalasaniN Nonalcoholic fatty liver disease and nonalcoholic steatohepatitis: Selected practical issues in their evaluation and managementHepatology200949130617DOI: 10.1002/hep.22603
[48]
KallwitzE. R McLachlanA CotlerS. J Role of peroxisome proliferators-activated receptors in the pathogenesis and treatment of nonalcoholic fatty liver diseaseWorld J Gastroenterol2008141228DOI: 10.3748/wjg.14.22
[49]
WuZ BucherN. L FarmerS. R Induction of peroxisome proliferator-activated receptor gamma during the conversion of 3T3 fibroblasts into adipocytes is mediated by C/EBPbeta, C/EBPdelta, and glucocorticoidsMol Cell Biol1996168412836DOI: 10.1128/mcb.16.8.4.128
[50]
FajasL FruchartJ. C AuwerxJ Transcriptional control of adipogenesisCurr Opin Cell Biol199810216573DOI: 10.1016/s0955-0674(98)80138-5
[51]
KimJ. B SpiegelmanB. M ADD1/SREBP1 promotes adipocyte differentiation and gene expression linked to fatty acid metabolismGenes Dev19961091096107DOI: 10.1101/gad.10.9.1.096
[52]
Schoonjans K Peinado-Onsurbe J Lefebvre A. M Heyman R. A Briggs M Deeb S Staels B Auwerx J PPARalpha and PPARgamma activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene EMBO J 1996 15 19 5336 48
[53]
FrohnertB. I HuiT. Y BernlohrD. A Identification of a functional peroxisome proliferator-responsive element in the murine fatty acid transport protein geneJ Biol Chem1999274739707DOI: 10.1074/jbc.274.7.3.970
[54]
YuJ. G JavorschiS HevenerA. L KruszynskaY. T NormanR. A SinhaM OlefskyJ. M The effect of thiazolidinediones on plasma adiponectin levels in normal, obese, and type 2 diabetic subjectsDiabetes20025110296874DOI: 10.2337/diabetes.51.10.2968
[55]
XuA WangY KeshawH XuL. Y LamK. S CooperG. J The fat-derived hormone adiponectin alleviates alcoholic and nonalcoholic fatty liver diseases in miceJ Clin Invest2003112191100DOI: 10.1172/JCI17797
[56]
SahaA. K AviluceaP. R YeJ. M AssifiM. M KraegenE. W RudermanN. B Pioglitazone treatment activates AMP-activated protein kinase in rat liver and adipose tissue in vivoBiochem Biophys Res Commun200431425805DOI: 10.1016/j.bbrc.2003.12.120
[57]
FryerL. G Parbu-PatelA CarlingD The Anti-diabetic drugs rosiglitazone and metformin stimulate AMP-activated protein kinase through distinct signaling pathwaysJ Biol Chem2002277282522632DOI: 10.1074/jbc. M202489200
[58]
GalliA CrabbD. W CeniE SalzanoR MelloT Svegliati-BaroniG RidolfiF TrozziL SurrentiC CasiniA Antidiabetic thiazolidinediones inhibit collagen synthesis and hepatic stellate cell activation in vivo and in vitroGastroenterology20021227192440DOI: 10.1053/gast.2002.33666
[59]
LeclercqI. A SempouxC StarkelP HorsmansY Limited therapeutic efficacy of pioglitazone on progression of hepatic fibrosis in ratsGut200655710209DOI: 10.1136/gut.2005.079194
[60]
MarraF EfsenE RomanelliR. G CaligiuriA PastacaldiS BatignaniG BonacchiA CaporaleR LaffiG PinzaniM GentiliniP Ligands of peroxisome proliferator-activated receptor gamma modulate profibrogenic and proinflammatory actions in hepatic stellate cellsGastroenterology2000119246678DOI: 10.1053/gast.2000.9365
[61]
BetteridgeD. J Effects of pioglitazone on lipid and lipoprotein metabolismDiabetes Obes Metab2007956407DOI: 10.1111/j.1463-1326.2007.00715.x
[62]
LincoffA. M WolskiK NichollsS. J NissenS. E Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes mellitus: a meta-analysis of randomized trialsJAMA20072981011808DOI: 10.1001/jama.298.10.1180
[63]
LokeY. K SinghS FurbergC. D Long-term use of thiazolidinediones and fractures in type 2 diabetes: a meta-analysisCMAJ20091801329DOI: 10.1503/cmaj.080486
[64]
NeumannA WeillA RicordeauP FagotJ. P AllaF AllemandH Pioglitazone and risk of bladder cancer among diabetic patients in France: a population-based cohort studyDiabetologia2012557195362DOI: 10.1007/s00125-012-2538-9
[65]
KirpichnikovD McFarlaneS. I MetforminJ. R. Sowers an updateAnn Intern Med200213712533DOI: 10.7326/0003-4819-137-1-200207020-00009
[66]
ZhouG MyersR LiY ChenY ShenX Fenyk-MelodyJ WuM VentreJ DoebberT FujiiN MusiN HirshmanM. F GoodyearL. J MollerD. E Role of AMP-activated protein kinase in mechanism of metformin actionJ Clin Invest20011088116774DOI: 10.1172/JCI13505
[67]
MadirajuA. K ErionD. M RahimiY ZhangX. M BraddockD. T AlbrightR. A PrigaroB. J WoodJ. L BhanotS MacDonaldM. J JurczakM. J CamporezJ. P LeeH. Y ClineG. W SamuelV. T KibbeyR. G ShulmanG. I Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenaseNature201451075065426DOI: 10.1038/nature13270
[68]
ColcaJ. R McDonaldW. G AdamsW. J MSDC-0602K, a metabolic modulator directed at the core pathology of non-alcoholic steatohepatitisExpert Opin Investig Drugs2018277631636DOI: 10.1080/13543784.2018.1494153
[69]
SodenJ. S DevereauxM. W HaasJ. E GumprichtE DahlR GrallaJ TraberM. G SokolR. J Subcutaneous vitamin E ameliorates liver injury in an in vivo model of steatocholestasisHepatology200746248595DOI: 10.1002/hep.21690
[70]
SokolR. J McKim Jr.J. M GoffM. C RuyleS. Z DevereauxM. W HanD PackerL EversonG Vitamin E reduces oxidant injury to mitochondria and the hepatotoxicity of taurochenodeoxycholic acid in the ratGastroenterology1998114116474DOI: 10.1016/s0016-5085(98)70644-4
[71]
AzziA GysinR KempnaP MunteanuA NegisY VillacortaL VisariusT ZinggJ. M Vitamin E mediates cell signaling and regulation of gene expressionAnn N Y Acad Sci200410318695DOI: 10.1196/annals.1331.009
[72]
MoranteM SandovalJ Gomez-CabreraM. C RodriguezJ. L PallardoF. V VinaJ. R TorresL BarberT Vitamin E deficiency induces liver nuclear factor-kappaB DNA-binding activity and changes in related genesFree Radic Res20053910112738DOI: 10.1080/10715760500193820
[73]
SanyalA. J ChalasaniN KowdleyK. V McCulloughA DiehlA. M BassN. M Neuschwander-TetriB. A LavineJ. E TonasciaJ UnalpA NattaM. Van ClarkJ BruntE. M KleinerD. E HoofnagleJ. H RobuckP. R NashC. R. N Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitisN Engl J Med201036218167585DOI: 10.1056/NEJMoa0907929
[74]
BjelakovicG NikolovaD GluudL. L SimonettiR. G GluudC Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysisJAMA2007297884257DOI: 10.1001/jama.297.8.8.42
[75]
SchurksM GlynnR. J RistP. M TzourioC KurthT Effects of vitamin E on stroke subtypes: meta-analysis of randomised controlled trialsBMJ2010341c5702DOI: 10.1136/bmj.c5702
[76]
KleinE. A Thompson Jr.I. M TangenC. M CrowleyJ. J LuciaM. S GoodmanP. J MinasianL. M FordL. G ParnesH. L GazianoJ. M KarpD. D LieberM. M WaltherP. J KlotzL ParsonsJ. K ChinJ. L DarkeA. K LippmanS. M GoodmanG. E Meyskens Jr.F. L BakerL. H Vitamin E and the risk of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial (SELECT)JAMA201130614154956DOI: 10.1001/jama.2011.1437
[77]
StrieterR. M RemickD. G WardP. A SpenglerR. N Lynch 3rdJ. P LarrickJ KunkelS. L Cellular and molecular regulation of tumor necrosis factor-alpha production by pentoxifyllineBiochem Biophys Res Commun1988155312306DOI: 10.1016/s0006-291x(88)81271-3
[78]
BhatV. B MadyasthaK. M Antioxidant and radical scavenging properties of 8-oxo derivatives of xanthine drugs pentoxifylline and lisofyllineBiochem Biophys Res Commun2001288512127DOI: 10.1006/bbrc.2001.5922
[79]
KoppeS. W SahaiA MalladiP WhitingtonP. F GreenR. M Pentoxifylline attenuates steatohepatitis induced by the methionine choline deficient dietJ Hepatol20044145928DOI: 10.1016/j.jhep.2004.06.030
[80]
ZeinC. O YerianL. M GogateP LopezR KirwanJ. P FeldsteinA. E McCulloughA. J Pentoxifylline improves nonalcoholic steatohepatitis: a randomized placebo-controlled trialHepatology201154516109DOI: 10.1002/hep.24544
[81]
BirkenfeldA. L ShulmanG. I Nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetesHepatology201459271323DOI: 10.1002/hep.26672
[82]
PuriP BaillieR. A WiestM. M MirshahiF ChoudhuryJ CheungO SargeantC ContosM. J SanyalA. J A lipidomic analysis of nonalcoholic fatty liver diseaseHepatology2007464108190DOI: 10.1002/hep.21763
[83]
LawitzE. J CosteA PoordadF AlkhouriN LooN McColganB. J TarrantJ. M NguyenT HanL ChungC RayA. S McHutchisonJ. G SubramanianG. M MyersR. P MiddletonM. S SirlinC LoombaR NyangauE FitchM LiK HellersteinM Acetyl-CoA Carboxylase Inhibitor GS-0976 for 12 Weeks Reduces Hepatic De Novo Lipogenesis and Steatosis in Patients With Nonalcoholic Steatohepatitis. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association, 16(12), 1983–19912018e3DOI: 10.1016/j.cgh.2018.04.042
[84]
StiedeK MiaoW BlanchetteH. S BeysenC HarrimanG HarwoodH. J. JR KelleyH KapellerR SchmalbachT WestlinWF Acetyl-coenzyme A carboxylase inhibition reduces de novo lipogenesis in overweight male subjects: A randomized, double-blind, crossover study. Hepatology,2017662324334DOI: 10.1002/hep.29246.
[85]
HodsonL FieldingB. A Stearoyl-CoA desaturase: rogue or innocent bystander?Prog Lipid Res20135211542DOI: 10.1016/j.plipres.2012.08.002
[86]
NtambiJ. M The regulation of stearoyl-CoA desaturase (SCD)Prog Lipid Res199534213950DOI: 10.1016/0163-7827(94)00010-j
[87]
MiyazakiM FlowersM. T SampathH ChuK OtzelbergerC LiuX NtambiJ. M Hepatic stearoyl-CoA desaturase-1 deficiency protects mice from carbohydrate-induced adiposity and hepatic steatosisCell Metab20076648496DOI: 10.1016/j.cmet.2007.10.014
[88]
IssandouM BouillotA BrusqJ. M ForestM. C GrillotD GuillardR MartinS MichielsC SulpiceT DauganA Pharmacological inhibition of stearoyl-CoA desaturase 1 improves insulin sensitivity in insulin-resistant rat modelsEur J Pharmacol20096181-32836DOI: 10.1016/j.ejphar.2009.07.004
[89]
WalleP TakkunenM MannistoV VaittinenM LankinenM KarjaV KakelaP AgrenJ TiainenM SchwabU KuusistoJ LaaksoM PihlajamakiJ Fatty acid metabolism is altered in non-alcoholic steatohepatitis independent of obesityMetabolism2016655655666DOI: 10.1016/j.metabol.2016.01.011
[90]
SafadiR KonikoffF. M MahamidM Zelber-SagiS HalpernM GilatT OrenR GroupF The fatty acid-bile acid conjugate Aramchol reduces liver fat content in patients with nonalcoholic fatty liver diseaseClin Gastroenterol Hepatol20141212208591 e1DOI: 10.1016/j.cgh.2014.04.038
[91]
EkstedtM FranzenL. E MathiesenU. L HolmqvistM BodemarG KechagiasS Statins in non-alcoholic fatty liver disease and chronically elevated liver enzymes: a histopathological follow-up studyJ Hepatol200747113541DOI: 10.1016/j.jhep.2007.02.013
[92]
TaylorF HuffmanM. D MacedoA. F MooreT. H BurkeM SmithG. Davey WardK EbrahimS Statins for the primary prevention of cardiovascular diseaseCochrane Database Syst Rev2013CD0048161DOI: 10.1002/14651858. CD004816.pub5
[93]
LonardoA BallestriS TargherG LoriaP Diagnosis and management of cardiovascular risk in nonalcoholic fatty liver diseaseExpert Rev Gastroenterol Hepatol20159562950DOI: 10.1586/17474124.2015.965143
[94]
LewisJ. H MortensenM. E ZweigS FuscoM. J MedoffJ. R BelderR StudyI. Pravastatin in Chronic Liver Disease Efficacy and safety of high-dose pravastatin in hypercholesterolemic patients with well-compensated chronic liver disease: Results of a prospective, randomized, double-blind, placebo-controlled, multicenter trialHepatology2007465145363DOI: 10.1002/hep.21848
[95]
AthyrosV. G TziomalosK GossiosT. D GrivaT AnagnostisP KargiotisK PagoureliasE. D TheocharidouE KaragiannisA MikhailidisD. P GroupG. S. C Safety and efficacy of long-term statin treatment for cardiovascular events in patients with coronary heart disease and abnormal liver tests in the Greek Atorvastatin and Coronary Heart Disease Evaluation (GREACE) Study: a post-hoc analysisLancet20103769756191622DOI: 10.1016/S0140-6736(10)61272-X
[96]
MinH. K KapoorA FuchsM MirshahiF ZhouH MaherJ KellumJ WarnickR ContosM. J SanyalA. J Increased hepatic synthesis and dysregulation of cholesterol metabolism is associated with the severity of nonalcoholic fatty liver diseaseCell Metab201215566574DOI: 10.1016/j.cmet.2012.04.004
[97]
JhaveriM. A KowdleyK. V New developments in the treatment of primary biliary cholangitis—Role of obeticholic acid. Ther. Clin. Risk Manag,20171310531060DOI: 10.2147/TCRM. S113052
[98]
BCariou The farnesoid X receptor (FXR) as a new target in non-alcoholic steatohepatitis. Diabetes Metab. 34(6 Pt 2),2008685691DOI: 10.1016/S1262-3636(08)74605-6
[99]
LefebvreP CariouB LienF KuipersF StaelsB Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev,2009891147191DOI: 10.1152/physrev.00010.2008.
[100]
KongB LuyendykJ. P TawfikO GuoG. L Farnesoid X receptor deficiency induces nonalcoholic steatohepatitis in low-density lipoprotein receptor-knockout mice fed a high-fat dietJ Pharmacol Exp Ther2009328111622DOI: 10.1124/jpet.108.144600
[101]
FiorucciS RizzoG AntonelliE RengaB MencarelliA RiccardiL OrlandiS PruzanskiM MorelliA PellicciariR A farnesoid x receptor-small heterodimer partner regulatory cascade modulates tissue metalloproteinase inhibitor-1 and matrix metalloprotease expression in hepatic stellate cells and promotes resolution of liver fibrosisJ Pharmacol Exp Ther2005314258495DOI: 10.1124/jpet.105.084905
[102]
DingL PangS SunY TianY YuL DangN Coordinated Actions of FXR and LXR in Metabolism: From Pathogenesis to Pharmacological Targets for Type 2 DiabetesInt J Endocrinol20142014751859DOI: 10.1155/2014/751859
[103]
RodaA PellicciariR GioielloA NeriF CamborataC PasseriD FrancoF. De SpinozziS CollivaC AdoriniL MontagnaniM AldiniR Semisynthetic bile acid FXR and TGR5 agonists: physicochemical properties, pharmacokinetics, and metabolism in the ratJ Pharmacol Exp Ther201435015668DOI: 10.1124/jpet.114.214650
[104]
PellicciariR CostantinoG CamaioniE SadeghpourB. M EntrenaA WillsonT. M FiorucciS ClericiC GioielloA Bile acid derivatives as ligands of the farnesoid X receptor. Synthesis, evaluation, and structure-activity relationship of a series of body and side chain modified analogues of chenodeoxycholic acidJ Med Chem20044718455969DOI: 10.1021/jm049904b
[105]
JhaveriM. A KowdleyK. V New developments in the treatment of primary biliary cholangitis - role of obeticholic acidTher Clin Risk Manag20171310531060DOI: 10.2147/TCRM. S113052
[106]
MudaliarS HenryR. R SanyalA. J MorrowL MarschallH. U KipnesM. AdoriniL SciaccaC. I CloptonP CastelloeE DillonP PruzanskiM ShapiroD Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology, 145(3), 574-5822013e1DOI: 10.1053/j.gastro.2013.05.042
[107]
RizzoG PasseriD FrancoF. De CiaccioliG DonadioL RizzoG OrlandiS SadeghpourB WangX. X JiangT LeviM PruzanskiM AdoriniL Functional characterization of the semisynthetic bile acid derivative INT-767, a dual farnesoid X receptor and TGR5 agonist. Mol Pharmacol.2010784617630DOI: 10.1124/mol.110.064501.
[108]
Neuschwander-TetriB. A LoombaR SanyalA. J LavineJ. E NattaM. L. Van AbdelmalekM. F ChalasaniN DasarathyS DiehlA. M HameedB KowdleyK. V McCulloughA TerraultN ClarkJ. M TonasciaJ BruntE. M KleinerD. E DooE NetworkN. C. R Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trialLancet2015385997295665DOI: 10.1016/S0140-6736(14)61933-4
[109]
RatziuV SanyalA. J LoombaR RinellaM HarrisonS AnsteeQ. M GoodmanZ BedossaP MacConellL ShringarpureR ShahA REGENERATEZ. Younossi Design of a pivotal, randomised, phase 3 study evaluating the safety and efficacy of obeticholic acid in patients with fibrosis due to nonalcoholic steatohepatitisContemp Clin Trials201984105803DOI: 10.1016/j.cct.2019.06.017
[110]
TullyD. C RuckerP. V ChianelliD WilliamsJ VidalA AlperP. B MutnickD BursulayaB SchmeitsJ WuX BaoD ZollJ KimY GroesslT McNamaraP SeidelH. M MolteniV LiuB PhimisterA JosephS. B LaffitteB Discovery of Tropifexor (LJN452), a Highly Potent Non-bile Acid FXR Agonist for the Treatment of Cholestatic Liver Diseases and Nonalcoholic Steatohepatitis (NASH)J Med Chem2017602499609973DOI: 10.1021/acs.jmedchem.7b00907
[111]
PedrosaM SeyedkazemiS FrancqueS SanyalA RinellaM CharltonM LoombaR RatziuV KochuparampilJ FischerL VaidyanathanS AnsteeQ. M A randomized, double-blind, multicenter, phase 2b study to evaluate the safety and efficacy of a combination of tropifexor and cenicriviroc in patients with nonalcoholic steatohepatitis and liver fibrosis: Study design of the TANDEM trialContemp Clin Trials202088105889DOI: 10.1016/j.cct.2019.105889
[112]
LamichaneS LamichaneB. Dahal KwonS. M Pivotal Roles of Peroxisome Proliferator-Activated Receptors (PPARs) and Their Signal Cascade for Cellular and Whole-Body Energy HomeostasisInt J Mol Sci2018194DOI: 10.3390/ijms19040949
[113]
KimH MendezR ZhengZ ChangL CaiJ ZhangR ZhangK Liver-enriched transcription factor CREBH interacts with peroxisome proliferator-activated receptor alpha to regulate metabolic hormone FGF21Endocrinology2014155376982DOI: 10.1210/en.2013-1490
[114]
VelkovT Interactions between Human Liver Fatty Acid Binding Protein and Peroxisome Proliferator Activated Receptor Selective DrugsPPAR Res20132013938401DOI: 10.1155/2013/938401
[115]
QuS SuD AltomonteJ KamagateA HeJ PerdomoG TseT JiangY DongH. H PPAR{alpha} mediates the hypolipidemic action of fibrates by antagonizing FoxO1Am J Physiol Endocrinol Metab20072922E42134DOI: 10.1152/ajpendo.00157.2006
[116]
MansouriR. M BaugeE StaelsB GervoisP Systemic and distal repercussions of liver-specific peroxisome proliferator-activated receptor-alpha control of the acute-phase responseEndocrinology20081496321523DOI: 10.1210/en.2007-1339
[117]
DeleriveP BosscherK. De BesnardS BergheW. Vanden PetersJ. M GonzalezF. J FruchartJ. C TedguiA HaegemanG StaelsB Peroxisome proliferator-activated receptor alpha negatively regulates the vascular inflammatory gene response by negative cross-talk with transcription factors NF-kappaB and AP-1J Biol Chem1999274453204854DOI: 10.1074/jbc.274.45.32048
[118]
NagasawaT InadaY NakanoS TamuraT TakahashiT MaruyamaK YamazakiY KurodaJ ShibataN Effects of bezafibrate, PPAR pan-agonist, and GW501516, PPARdelta agonist, on development of steatohepatitis in mice fed a methionine- and choline-deficient dietEur J Pharmacol20065361-218291DOI: 10.1016/j.ejphar.2006.02.028
[119]
RatziuV HarrisonS. A FrancqueS BedossaP LehertP SerfatyL Romero-GomezM BoursierJ AbdelmalekM CaldwellS DrenthJ AnsteeQ. M HumD HanfR RoudotA MegnienS StaelsB SanyalA GroupG.-I. S Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-alpha and -delta, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis WorseningGastroenterology2016150511471159 e5DOI: 10.1053/j.gastro.2016.01.038
[120]
ACCardoso Cde Figueiredo-Mendes CAVillela-Nogueira AJSanyal New drugs for non-alcoholic steatohepatitisLiver Int202040196101DOI: 10.1111/liv.14354
[121]
SinhaR. A BruinstroopE SinghB. K YenP. M Nonalcoholic Fatty Liver Disease and Hypercholesterolemia: Roles of Thyroid Hormones, Metabolites, and AgonistsThyroid201929911731191DOI: 10.1089/thy.2018.0664
[122]
BohincB. N MichelottiG XieG PangH SuzukiA GuyC. D PiercyD KrugerL Swiderska-SynM MachadoM PereiraT ZavackiA. M AbdelmalekM DiehlA. M Repair-related activation of hedgehog signaling in stromal cells promotes intrahepatic hypothyroidismEndocrinology2014155114591601DOI: 10.1210/en.2014-1302
[123]
SinhaR. A BruinstroopE SinghB. K YenP. M Thyroid hormones and thyromimetics: A new approach to NASH?Hepatology2020DOI: 10.1002/hep.31204
[124]
KellyM. J Pietranico-ColeS LariganJ. D HaynesN. E ReynoldsC. H ScottN VermeulenJ DvorozniakM Conde-KnapeK HuangK. S SoS. S ThakkarK QianY BannerB MennonaF DanziS KleinI TaubR TilleyJ Discovery of 2-(3,5-dichloro-4-(5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yloxy)phenyl)-3,5-dio xo-2,3,4,5-tetrahydro(1,2,4)triazine-6-carbonitrile (MGL-3196), a Highly Selective Thyroid Hormone Receptor beta agonist in clinical trials for the treatment of dyslipidemiaJ Med Chem20145710391223DOI: 10.1021/jm4019299
[125]
HarrisonS. A BashirM. R GuyC. D ZhouR MoylanC. A FriasJ. P AlkhouriN BansalM. B BaumS Neuschwander-TetriB. A TaubR MoussaS. E Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet,20193941021320122024DOI: 10.1016/S0140-6736(19)32517-6.
[126]
LoombaR NeutelJ MohseniR BernardD SeveranceR DaoM SainiS MargaritescuC HomerK TranB ManciniM MasamuneH LianB VK2809, a novel liver-directed thyroid receptor beta agonist, significantly reduces liver fat with both low and high doses in patients with non-alcoholic fatty liver disease: a phase 2 randomized, placebo-controlled trial. J Hepatol,201970e150151DOI: 10.1016/S0618-8278(19)30266-X
[127]
JinnouchiH SugiyamaS YoshidaA HieshimaK KurinamiN SuzukiT MiyamotoF KajiwaraK MatsuiK JinnouchiT Liraglutide, a glucagon-like peptide-1 analog, increased insulin sensitivity assessed by hyperinsulinemic-euglycemic clamp examination in patients with uncontrolled type 2 diabetes mellitusJ Diabetes Res20152015706416DOI: 10.1155/2015/706416
[128]
BernsmeierC Meyer-GerspachA. C BlaserL. S JekerL SteinertR. E HeimM. H BeglingerC Glucose-induced glucagon-like Peptide 1 secretion is deficient in patients with non-alcoholic fatty liver diseasePLoS One201491e87488DOI: 10.1371/journal.pone.0087488
[129]
DruckerD. J NauckM. A The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetesLancet200636895481696705DOI: 10.1016/S0140-6736(06)69705-5
[130]
DhirG CusiK Glucagon like peptide-1 receptor agonists for the management of obesity and non-alcoholic fatty liver disease: a novel therapeutic optionJ Investig Med2018661710DOI: 10.1136/jim-2017-000554
[131]
WangX. C GusdonA. M LiuH QuS Effects of glucagon-like peptide-1 receptor agonists on non-alcoholic fatty liver disease and inflammationWorld J Gastroenterol201420401482130DOI: 10.3748/wjg.v20.i40.14821
[132]
ArmstrongM. J GauntP AithalG. P BartonD HullD ParkerR HazlehurstJ. M GuoK teamL. t AboudaG AldersleyM. A StockenD GoughS. C TomlinsonJ. W BrownR. M HubscherS. G NewsomeP. N Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 studyLancet20163871001967990DOI: 10.1016/S0140-6736(15)00803-X
[133]
TraceyK. J CeramiA Tumor necrosis factor, other cytokines and diseaseAnnu Rev Cell Biol1993931743DOI: 10.1146/annurev.cb.09.110193.001533
[134]
HotamisligilG. S ShargillN. S SpiegelmanB. M Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistanceScience199325950918791DOI: 10.1126/science.7678183
[135]
UysalK. T WiesbrockS. M MarinoM. W HotamisligilG. S Protection from obesity-induced insulin resistance in mice lacking TNF-alpha functionNature199738966516104DOI: 10.1038/39335
[136]
MoschenA. R MolnarC GeigerS GraziadeiI EbenbichlerC. F WeissH KaserS KaserA TilgH Anti-inflammatory effects of excessive weight loss: potent suppression of adipose interleukin 6 and tumour necrosis factor alpha expressionGut2010599125964DOI: 10.1136/gut.2010.214577
[137]
DandonaP WeinstockR ThusuK Abdel-RahmanE AljadaA WaddenT Tumor necrosis factor-alpha in sera of obese patients: fall with weight lossJ Clin Endocrinol Metab1998838290710DOI: 10.1210/jcem.83.8.5.026
[138]
KernP. A SaghizadehM OngJ. M BoschR. J DeemR SimsoloR. B The expression of tumor necrosis factor in human adipose tissue. Regulation by obesity, weight loss, and relationship to lipoprotein lipaseJ Clin Invest199595521119DOI: 10.1172/JCI117899
[139]
HoofM. Van RahierJ HorsmansY Tamoxifen-induced steatohepatitisAnn Intern Med199612498556DOI: 10.7326/0003-4819-124-9-199605010-00015
[140]
ValentiL FracanzaniA. L DongiovanniP SantorelliG BranchiA TaioliE FiorelliG FargionS Tumor necrosis factor alpha promoter polymorphisms and insulin resistance in nonalcoholic fatty liver diseaseGastroenterology2002122227480DOI: 10.1053/gast.2002.31065
[141]
LdeF. Pinto CompriC. M FornariJ. V BartchewskyW CintraD. E TrevisanM PdeO. Carvalho RibeiroM. L VellosoL. A SaadM. J Pedrazzoli Jr.J GamberoA The immunosuppressant drug, thalidomide, improves hepatic alterations induced by a high-fat diet in miceLiver Int201030460310DOI: 10.1111/j.1478-3231.2009.02200.x
[142]
KocaS. S BahceciogluI. H PoyrazogluO. K OzercanI. H SahinK UstundagB The treatment with antibody of TNF-alpha reduces the inflammation, necrosis and fibrosis in the non-alcoholic steatohepatitis induced by methionine- and choline-deficient dietInflammation2008312918DOI: 10.1007/s10753-007-9053-z
[143]
LiZ YangS LinH HuangJ WatkinsP. A MoserA. B DesimoneC SongX. Y DiehlA. M Probiotics and antibodies to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver diseaseHepatology200337234350DOI: 10.1053/jhep.2003.50048
[144]
LesmanaC. R HasanI BudihusodoU GaniR. A KrisnuhoniE AkbarN LesmanaL. A Diagnostic value of a group of biochemical markers of liver fibrosis in patients with non-alcoholic steatohepatitisJ Dig Dis20091032016DOI: 10.1111/j.1751-2980.2009.00386.x
[145]
CrespoJ CayonA Fernandez-GilP Hernandez-GuerraM MayorgaM Dominguez-DiezA Fernandez-EscalanteJ. C Pons-RomeroF Gene expression of tumor necrosis factor alpha and TNF-receptors, p55 and p75, in nonalcoholic steatohepatitis patientsHepatology2001346115863DOI: 10.1053/jhep.2001.29628
[146]
HuiJ. M HodgeA FarrellG. C KenchJ. G KriketosA GeorgeJ Beyond insulin resistance in NASH: TNF-alpha or adiponectin?Hepatology20044014654DOI: 10.1002/hep.20280
[147]
MullerS MartinS KoenigW Hanifi-MoghaddamP RathmannW HaastertB GianiG IlligT ThorandB KolbH Impaired glucose tolerance is associated with increased serum concentrations of interleukin 6 and co-regulated acute-phase proteins but not TNF-alpha or its receptorsDiabetologia200245680512DOI: 10.1007/s00125-002-0829-2
[148]
BruunJ. M VerdichC ToubroS AstrupA RichelsenB Association between measures of insulin sensitivity and circulating levels of interleukin-8, interleukin-6 and tumor necrosis factor-alpha. Effect of weight loss in obese menEur J Endocrinol2003148553542DOI: 10.1530/eje.0.1480535
[149]
DouglasH. E TGF-ss in wound healing: a reviewJ Wound Care20101994036DOI: 10.12968/jowc.2010.19.9.7.8235
[150]
FriedmanS. L Cytokines and fibrogenesisSemin Liver Dis199919212940DOI: 10.1055/s-2007-1007105
[151]
ShekF. W BenyonR. C How can transforming growth factor beta be targeted usefully to combat liver fibrosis?Eur J Gastroenterol Hepatol20041621236DOI: 10.1097/00042737-200402000-00001
[152]
BissellD. M RoulotD GeorgeJ Transforming growth factor beta and the liverHepatology200134585967DOI: 10.1053/jhep.2001.28457
[153]
BleserP. J. De NikiT RogiersV GeertsA Transforming growth factor-beta gene expression in normal and fibrotic rat liverJ Hepatol199726488693DOI: 10.1016/s0168-8278(97)80257-7
[154]
HellerbrandC StefanovicB GiordanoF BurchardtE. R BrennerD. A The role of TGFbeta1 in initiating hepatic stellate cell activation in vivoJ Hepatol19993017787DOI: 10.1016/s0168-8278(99)80010-5
[155]
CzajaM. J WeinerF. R FlandersK. C GiambroneM. A WindR BiempicaL ZernM. A In vitro and in vivo association of transforming growth factor-beta 1 with hepatic fibrosisJ Cell Biol19891086247782DOI: 10.1083/jcb.108.6.2.477
[156]
AnnoniG WeinerF. R ZernM. A Increased transforming growth factor-beta 1 gene expression in human liver diseaseJ Hepatol1992142-325964DOI: 10.1016/0168-8278(92)90168-o
[157]
CastillaA PrietoJ FaustoN Transforming growth factors beta 1 and alpha in chronic liver disease. Effects of interferon alfa therapyN Engl J Med19913241493340DOI: 10.1056/NEJM199104043241401
[158]
Milani S Herbst H Schuppan D Stein H Surrenti C Transforming growth factors beta 1 and beta 2 are differentially expressed in fibrotic liver disease Am J Pathol 1991 139 6 1221 9
[159]
StarkelP SempouxC LeclercqI HerinM DebyC DesagerJ. P HorsmansY Oxidative stress, KLF6 and transforming growth factor-beta up-regulation differentiate non-alcoholic steatohepatitis progressing to fibrosis from uncomplicated steatosis in ratsJ Hepatol200339453846DOI: 10.1016/s0168-8278(03)00360-x
[160]
HasegawaT YonedaM NakamuraK MakinoI TeranoA Plasma transforming growth factor-beta1 level and efficacy of alpha-tocopherol in patients with non-alcoholic steatohepatitis: a pilot studyAliment Pharmacol Ther20011510166772DOI: 10.1046/j.1365-2036.2001.01083.x
[161]
FabregatI Moreno-CàceresJ SánchezA DooleyS DewidarB GiannelliG DijkeP. ten TGF-β signalling and liver disease. FEBS J,201628322192232DOI: 10.1111/febs.13665
[162]
LuangmonkongT SurigugaS BigaevaE BoersemaM OosterhuisD JongK. P. de SchuppanD MutsaersH. A. M OlingaP Evaluating the antifibrotic potency of galunisertib in a human ex vivo model of liver fibrosis. Br. J. Pharmacol.20171741831073117DOI: 10.1111/bph.13945
[163]
DerynckR ZhangY. E Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature,20034256958577584DOI: 10.1038/nature02006
[164]
FabregatI FernandoJ MainezJ SanchoP TGF-beta signaling in cancer treatment. Curr. Pharm. Des.2014201729342947DOI: 10.2174/13816128113199990591
[165]
ELefebvre GMoyle RReshef LPRichman MThompson FHong HLChou THashiguchi CPlato DPoulin TRichards HYoneyama HJenkins GWolfgang SLFriedman Antifibrotic Effects of the Dual CCR2/CCR5 AntagonistCenicrivirocin Animal Models of Liver and Kidney Fibrosis. PLoS One. Jun 272016116e0158156DOI: 10.1371/journal.pone.0158156.
[166]
FriedmanS SanyalA GoodmanZ LefebvreE GottwaldM FischerL RatziuV Efficacy and safety study of cenicriviroc for the treatment of non-alcoholic steatohepatitis in adult subjects with liver fibrosis: CENTAUR Phase 2b study design. Contemp Clin Trials,201647356365DOI: 10.1016/j.cct.2016.02.012
[167]
MederackeI HsuC. C TroegerJ. S HuebenerP MuX DapitoD. H PradereJ. P SchwabeR. F Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiologyNat Commun201342823DOI: 10.1038/ncomms3823
[168]
FriedmanS. L Molecular regulation of hepatic fibrosis, an integrated cellular response to tissue injuryJ Biol Chem20002754224750DOI: 10.1074/jbc.275.4.2.247
[169]
HigashiT FriedmanS. L HoshidaY Hepatic stellate cells as key target in liver fibrosisAdv Drug Deliv Rev20171212742DOI: 10.1016/j.addr.2017.05.007
[170]
SchaferS ViswanathanS WidjajaA. A LimW. W Moreno-MoralA DeLaughterD. M NgB PatoneG ChowK KhinE TanJ ChothaniS. P YeL RackhamO. J. L KoN. S. J SahibN. E PuaC. J ZhenN. T. G XieC WangM MaatzH LimS SaarK BlachutS PetrettoE SchmidtS PutoczkiT Guimaraes-CamboaN WakimotoH HeeschS. van SigmundssonK LimS. L SoonJ. L ChaoV. T. T ChuaY. L TanT. E EvansS. M LohY. J JamalM. H OngK. K ChuaK. C OngB. H ChakaramakkilM. J SeidmanJ. G SeidmanC. E HubnerN SinK. Y. K CookS. A IL-11 is a crucial determinant of cardiovascular fibrosisNature20175527683110115DOI: 10.1038/nature24676
[171]
NgB DongJ D'AgostinoG ViswanathanS WidjajaA. A LimW. W KoN. S. J TanJ ChothaniS. P HuangB XieC PuaC. J ChackoA. M Guimaraes-CamboaN EvansS. M ByrneA. J MaherT. M LiangJ JiangD NobleP. W SchaferS CookS. A Interleukin-11 is a therapeutic target in idiopathic pulmonary fibrosisSci Transl Med201911511DOI: 10.1126/scitranslmed.aaw1237
[172]
WidjajaA. A SinghB. K AdamiE ViswanathanS DongJ D'AgostinoG. A NgB LimW. W TanJ PalejaB. S TripathiM LimS. Y ShekeranS. G ChothaniS. P RabesA SombetzkiM BruinstroopE MinL. P SinhaR. A AlbaniS YenP. M SchaferS CookS. A Inhibiting Interleukin 11 Signaling Reduces Hepatocyte Death and Liver Fibrosis, Inflammation, and Steatosis in Mouse Models of Nonalcoholic SteatohepatitisGastroenterology20191573777792 e14DOI: 10.1053/j.gastro.2019.05.002
[173]
KleinS MittendorferB EagonJ. C PattersonB GrantL FeirtN SekiE BrennerD KorenblatK McCreaJ Gastric bypass surgery improves metabolic and hepatic abnormalities associated with nonalcoholic fatty liver diseaseGastroenterology20061306156472DOI: 10.1053/j.gastro.2006.01.042
[174]
VianaE. C Araujo-DasilioK. L MiguelG. P BressanJ LemosE. M MoysesM. R AbreuG. R. de AzevedoJ. L. de CarvalhoP. S Passos-BuenoM. R ErreraF. I BissoliN. S Gastric bypass and sleeve gastrectomy: the same impact on IL-6 and TNF-alpha. Prospective clinical trialObes Surg2013238125261DOI: 10.1007/s11695-013-0894-2
[175]
Gastrointestinal surgery for severe obesity. NIH consensus development conference, March 25-7,1991 Nutrition 1996 12 6 397 404
[176]
BurgueraB AgustiA ArnerP BaltasarA BarbeF BarceloA BretonI CabanesT CasanuevaF. F CouceM. E DieguezC FiolM RealJ. M. Fernandez FormigueraX FruhbeckG RomeroM. Garcia SanzM. Garcia GhigoE GomisR HigaK IbarraO LacyA LarradA MasmiquelL MoizeV MorenoB MoreiroJ RicartW RiescoM SalinasR SalvadorJ Pi-SunyerF. X ScopinaroN SjostromL PaganA PeregV PernauteA. Sanchez TorresA UrgelesJ. R Vidal-PuigA VidalJ VilaM Critical assessment of the current guidelines for the management and treatment of morbidly obese patientsJ Endocrinol Invest2007301084452DOI: 10.1007/bf03349226
[177]
BowerG TomaT HarlingL JiaoL. R EfthimiouE DarziA AthanasiouT SurgeryH. Ashrafian: Bariatric DiseaseNon-Alcoholic Fatty Liver a Systematic Review of Liver Biochemistry and HistologyObes Surg2015251222809DOI: 10.1007/s11695-015-1691-x
[178]
Chavez-TapiaN. C Tellez-AvilaF. I Barrientos-GutierrezT Mendez-SanchezN Lizardi-CerveraJ UribeM Bariatric surgery for non-alcoholic steatohepatitis in obese patientsCochrane Database Syst Rev2010CD0073401DOI: 10.1002/14651858. CD007340.pub2
[179]
RouxC. W. le HeneghanH. M Bariatric Surgery for ObesityMed Clin North Am20181021165182DOI: 10.1016/j.mcna.2017.08.011
[180]
TohS. Y ZarshenasN JorgensenJ Prevalence of nutrient deficiencies in bariatric patientsNutrition20092511-1211506DOI: 10.1016/j.nut.2009.03.012
[181]
ParrottJ FrankL RabenaR Craggs-DinoL IsomK. A GreimanL American Society for Metabolic and Bariatric Surgery Integrated Health Nutritional Guidelines for the Surgical Weight Loss Patient 2016 Update: MicronutrientsSurg Obes Relat Dis2017135727741DOI: 10.1016/j.soard.2016.12.018
[182]
HammerH. F Medical complications of bariatric surgery: focus on malabsorption and dumping syndromeDig Dis20123021826DOI: 10.1159/000336681
[183]
MarskR JonasE RasmussenF NaslundE Nationwide cohort study of post-gastric bypass hypoglycaemia including 5,040 patients undergoing surgery for obesity in 1986-2006 in SwedenDiabetologia20105311230711DOI: 10.1007/s00125-010-1798-5
[184]
LiX LiC Analysis of changes in intestinal flora and intravascular inflammation and coronary heart disease in obese patientsExp Ther Med201815545384542DOI: 10.3892/etm.2018.5987
[185]
LiC. Y DempseyJ. L WangD LeeS WeigelK. M FeiQ BhattD. K PrasadB RafteryD GuH CuiJ. Y PBDEs Altered Gut Microbiome and Bile Acid Homeostasis in Male C57BL/6 MiceDrug Metab Dispos201846812261240DOI: 10.1124/dmd.118.081547
[186]
VahedS. Zununi SaniH. Moghaddas SaadatY. Rahbar BarzegariA OmidiY Type 1 diabetes: Through the lens of human genome and metagenome interplayBiomed Pharmacother2018104332342DOI: 10.1016/j.biopha.2018.05.052
[187]
ParkJ. S SeoJ. H YounH. S Gut microbiota and clinical disease: obesity and nonalcoholic Fatty liver diseasePediatr Gastroenterol Hepatol Nutr2013161227DOI: 10.5223/pghn.2013.16.1.2.2
[188]
QinJ LiR RaesJ ArumugamM BurgdorfK. S ManichanhC NielsenT PonsN LevenezF YamadaT MendeD. R LiJ XuJ LiS LiD CaoJ WangB LiangH ZhengH XieY TapJ LepageP BertalanM BattoJ. M HansenT PaslierD. Le LinnebergA NielsenH. B PelletierE RenaultP Sicheritz-PontenT TurnerK ZhuH YuC LiS JianM ZhouY LiY ZhangX LiS QinN YangH WangJ BrunakS DoreJ GuarnerF KristiansenK PedersenO ParkhillJ WeissenbachJ MetaH. I. T. C BorkP EhrlichS. D WangJ A human gut microbial gene catalogue established by metagenomic sequencingNature201046472855965DOI: 10.1038/nature08821
[189]
SantacruzA ColladoM. C Garcia-ValdesL SeguraM. T Martin-LagosJ. A AnjosT Marti-RomeroM LopezR. M FloridoJ CampoyC SanzY Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant womenBr J Nutr201010418392DOI: 10.1017/S0007114510000176
[190]
Feitoza A. Baddini Pereira A. Fernandes Costa N. Ferreira da Ribeiro B. Goncalves Conjugated linoleic acid (CLA): effect modulation of body composition and lipid profile Nutr Hosp 2009 24 4 422 8
[191]
ZhangC BjorkmanA CaiK LiuG WangC LiY XiaH SunL KristiansenK WangJ HanJ HammarstromL Pan-HammarstromQ Impact of a 3-Months Vegetarian Diet on the Gut Microbiota and Immune RepertoireFront Immunol20189908DOI: 10.3389/fimmu.2018.00908
[192]
ShinN. R WhonT. W ProteobacteriaJ. W. Bae microbial signature of dysbiosis in gut microbiotaTrends Biotechnol2015339496503DOI: 10.1016/j.tibtech.2015.06.011
[193]
LeungC RiveraL FurnessJ. B AngusP. W The role of the gut microbiota in NAFLDNat Rev Gastroenterol Hepatol201613741225DOI: 10.1038/nrgastro.2016.85
[194]
BeleiO OlariuL DobrescuA MarcoviciT MargineanO The relationship between non-alcoholic fatty liver disease and small intestinal bacterial overgrowth among overweight and obese children and adolescentsJ Pediatr Endocrinol Metab2017301111611168DOI: 10.1515/jpem-2017-0252
[195]
AlvesC. C WaitzbergD. L AndradeL. S. de AguiarL. Dos Santos ReisM. B GuanabaraC. C JuniorO. A RibeiroD. A SalaP Prebiotic and Synbiotic Modifications of Beta Oxidation and Lipogenic Gene Expression after Experimental Hypercholesterolemia in Rat LiverFront Microbiol201782010DOI: 10.3389/fmicb.2017.02010
[196]
BakkerG. J TransplantationM. Nieuwdorp: Fecal Microbiota Therapeutic Potential for a Multitude of Diseases beyond Clostridium difficileMicrobiol Spectr201754DOI: 10.1128/microbiolspec. BAD-0008-2017
[197]
GrootP. F. de FrissenM. N ClercqN. C. de NieuwdorpM Fecal microbiota transplantation in metabolic syndrome: History, present and futureGut Microbes201783253267DOI: 10.1080/19490976.2017.1293224
[198]
WoodhouseC. A PatelV. C SinganayagamA ShawcrossD. L Review article: the gut microbiome as a therapeutic target in the pathogenesis and treatment of chronic liver diseaseAliment Pharmacol Ther2018472192202DOI: 10.1111/apt.14397
[199]
ZhouD PanQ ShenF CaoH. X DingW. J ChenY. W FanJ. G Total fecal microbiota transplantation alleviates high-fat diet-induced steatohepatitis in mice via beneficial regulation of gut microbiotaSci Rep2017711529DOI: 10.1038/s41598-017-01751-y
[200]
RoyT. Le LlopisM LepageP BruneauA RabotS BevilacquaC MartinP PhilippeC WalkerF BadoA PerlemuterG Cassard-DoulcierA. M GerardP Intestinal microbiota determines development of non-alcoholic fatty liver disease in miceGut20136212178794DOI: 10.1136/gutjnl-2012-303816
[201]
VriezeA NoodE. Van HollemanF SalojarviJ KootteR. S BartelsmanJ. F Dallinga-ThieG. M AckermansM. T SerlieM. J OozeerR DerrienM DruesneA VliegJ. E. Van Hylckama BloksV. W GroenA. K HeiligH. G ZoetendalE. G StroesE. S VosW. M. de HoekstraJ. B NieuwdorpM Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndromeGastroenterology201214349136 e7DOI: 10.1053/j.gastro.2012.06.031
Share
Back to top