Edanur Topal1, Kader Aydemir1, Özge Çağlar2,3, Begüm Arda1, Onur Kayabaşı1, Manolya Yıldız1, Elif Özyılmaz3, Oytun Erbaş1

1ERBAS Institute of Experimental Medicine, Illinois, USA & Gebze, Turkey
2Selçuk University, Faculty of Science, Department of Chemistry, Konya/Turkey
3Selçuk University, Faculty of Science, Department of Biochemistry, Konya/Turkey

Keywords: Alcohol, alcoholic fatty liver disease, fructose, liver, non-alcoholic fatty liver disease

Abstract

The liver is the largest organ of our body, which plays a role in the physiological process in the body and is most exposed to toxins. The liver's functions are essential for life since it is involved in macronutrient metabolism. Fatty diets, excessive alcohol consumption, sedentary lifestyles, ready-made foods and beverages, and fructose metabolism in the liver are all factors that contribute to liver fat. In the advanced stage of fatty liver, changes in liver cells, cirrhosis, and hepatocellular carcinoma, which rank first in the world in terms of mortality, are seen. Therefore, current treatments and future approaches to fatty liver research are of tremendous interest. The results of investigations looking into the causes and current treatments for alcoholic liver disease and non-alcoholic fatty liver disease were compiled in this review.

The liver is the body's largest organ and is essential for maintaining the body's balance. Therefore, it is a vital organ for life, as it is responsible for the entire body's metabolism and participates in a variety of crucial physiological processes.[1-3] The breakdown of xenobiotic compounds, the realization of gluconeogenic reactions, the production of bile, the oxidation of lipids, the supply of energy for the continuation of physiological processes, the detoxification, digestion, and metabolism of the blood. In addition, the metabolism of amino acids and the removal of nitrogenous wastes are all handled by this organ. It also plays a crucial function in metabolism, protein synthesis, and amino acid metabolism.[4-6] Viral hepatitis, alcohol consumption, sedentary life, hepatic fat accumulation of 5% or more, drug-induced liver disease, autoimmune liver disease, obesity, Type 2 diabetes, and metabolic syndromes, such as high-fat diet, corn syrup, high-fructose foods, and pizza excessive use of quick meals and sugary drinks cause fatty liver.[7-12] There are two main types of fatty liver disease: alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD). The hepatic portal vein transports the majority of the blood to the liver from the spleen and gastrointestinal system. This venous blood contains antigens that are then exposed to the immune repertory of the liver, these antigens establishing the liver as a critical immunological site for both immune tolerance and activation. Irregular immunological processes can affect both the liver and systemic immune responses. Kupffer cells, mucosal-associated invariant T (MAIT) cells, αβ T cells, lymphoid cells (ILCs), γδ T cells, natural killer T (NKT) cells, and natural killer (NK) cells all reside in liver B cells.[4,13] NK cells make up 40% of the total lymphocytes in the human liver.[4,14] This result shows that NK cells have an important potential in the regulation of immunity in the liver.[4] Depending on the type of liver damage and the underlying causes, immune reactions are affected by a variety of mechanisms.[15,16] Chronic inflammation has been indicated as an important trigger of liver fibrogenesis.[17]

FATTY LIVER AND INFLAMMATION

Various defense systems have evolved in living things in response to attacks on cells and tissues. Inflammation is defined as a physiological response to tissue damage when the organism is exposed to biological, physical, or chemical factors.[18,19] Cytokines are cellular regulatory proteins. Special cells (T lymphocytes) release them in response to specific stimuli, and they influence the behavior of targeted cells. The effects of cytokines might be systemic or local. Although different cells in different tissues secrete the same cytotoxin, it has the same biological function.[20] Kupffer cells generate a considerable amount of inflammatory factors when the liver is wounded. To treat and prevent liver damage induced by alcohol intake, it is vital to limit the liver's inflammatory response.[21] Metabolic syndrome studies have reported that patients have increased inflammation, which facilitates the development of fatty liver, and increases the value of some inflammatory mediators, including tumor necrosis factor-alpha (TNF-α) and lipid peroxidase, in fatty liver.[22,23] In the pathogenesis of ALD and NAFLD, a high number of inflammatory cells and inflammatory mediators play both useful and destructive roles in the liver. It causes hepatocyte destruction and liver fibrosis, for example, while also accelerating liver regeneration and protecting the liver from bacterial infections. Inflammation generated by low bacterial infection causes liver damage at both the ALD and the NAFLD, where liver regeneration is not required due to mild harm.[24] Another inflammatory cytokine, interleukin-22 (IL-22), promotes liver regeneration, whereas interleukin-17 (IL-17) induces liver injury. In response, the expression profiles of antagonistic cytokines shift towards IL-17 in the advanced stage.[25] Cytokines also have an important role in the development of NAFLD.[26] TNF-α is an inflammatory cytokine produced by a variety of cells, including macrophages and kupffer cells in the liver. TNF-α is a crucial factor in the development of inflammation and insulin resistance.[27]

ALCOHOLIC LIVER DISEASE

Alcoholic liver disease is a term used to describe liver disease caused by excessive alcohol use. Lipoatrophy starts with fat buildup in the liver and can progress to severe alcoholic steatohepatitis.[28] When they are detected early and their alcohol use is minimized, some persons may be recycled.[29]

Ethanol (EtOH) is produced by the enzymatic action of brewer's yeast on sugar-containing compounds during fermentation or distillation.[30] GABAergic neurotransmission is harmed by long-term use of EtOH. Oxidative stress, astrocytic dysfunction, neuroinflammation, and neurotoxicity are all caused by chronic EtOH exposure.[31] The stomach absorbs 20% of ethanol quickly, whereas the duodenum absorbs the remaining 80%.[32,33] Because ingested alcohol dissolves quickly in water, it enters the bloodstream and is transported to the tissues immediately. Alcohol, which is easily swallowed and passes through the mucosal epithelium, begins to be absorbed before it reaches the stomach due to its short-chain chemical structure. The rate of alcohol absorption is inversely related to stomach fullness and directly proportional to the rate of alcohol consumption. While only 5-8% of the alcohol consumed by the circulatory system is expelled from the body without being processed through respiration, urine, or perspiration, enters 90% is destruction process in the liver.[30] In the liver, there are two phases to the breakdown of alcohol. The oxidation of ethanol to acetaldehyde and the conversion of acetaldehyde to acetic acid are the two stages involved. In oxidation, both the alcohol dehydrogenase route and the microsomal ethanol oxidation system are involved. The microsomal ethanol-oxidizing system (MEOS) or the cytochrome P450 family 2 subfamily E member 1 (CYP2E1) enzymes are both involved. Alcohol dehydrogenase is a zinc-containing cytosolic enzyme found in the cytoplasm of liver cells. Almost all of the conversion takes place through this pathway when a low dose of alcohol is consumed. The liver's NAD+ capacity restricts the rate of breakdown to 8-10 g ethanol/ hour. Hunger and a low-protein diet slow the breakdown of alcohol. The CYP2E1 enzyme is present in the endoplasmic reticulum of the liver. Because the enzyme's activity is 4-40 times lower than that of alcohol dehydrogenase, it has no importance on the breakdown of alcohol in modest or moderate quantities. When the liver's NAD+ capacity is exceeded (one promille = alcohol level greater than 100 mg/ dl), it becomes increasingly significant. Regular use of alcohol activates the CYP2E1 enzyme. (enzyme efficacy may increase up to ten times). Increased enzyme content accelerates the deterioration rate of alcohol and decreases blood levels.[34,35] Most of the acetaldehyde is oxidized by the acetaldehyde dehydrogenase 2 (ALDH2) variant.[36] Slow inactivator individuals who are homozygous or heterozygous for low activity have an acetaldehyde response, which causes consequences like blushing even when eaten in little doses. These people are more numerous in East Asian races and have a lower risk of ALD.[37] It is defined in American dietary guidelines as "one drink containing approximately 14 g of alcohol". This ratio corresponds to approximately 150 ml of wine (12-13% weight/volume), 350 ml of beer (5% weight/volume), or 40-45 ml of liqueur (40-45% weight/volume).[38]

The National Institute on Alcohol Abuse and Alcoholism (NIAAA) defines alcohol use disorder as more than two drinks per day for women and more than three drinks per day for men, while excess alcohol intake is specified as four for women and more than five for men within two hours.[39] Individuals with an alcohol use disorder may be diagnosed with alcoholic fatty liver disease (AFLD). Alcohol-induced liver steatosis, which is generally seen in the form of macrovesicular steatosis, is seen in approximately 90% of individuals with high alcohol consumption and may develop within two weeks after continuous and heavy alcohol consumption. If alcohol is completely abandoned, improvements in lipolysis are observed.[40]

NON-ALCOHOLIC FATTY LIVER DISEASE

Non-alcoholic fatty liver disease is now known as metabolic dysfunction-associated fatty liver disease (MAFLD).[41,42] It is stated that the disease affects approximately one-quarter of the world population and it is associated with significant morbidity and mortality.[42-44] A recent study predicts that NAFLD-related liver diseases will increase from 64% to 156% worldwide by 2030.[45] Obese people have a rate of 90%, Type 2 diabetics have a rate of 76%, and non-overweight people have a rate of 16%.[46,47] In the coming years, NAFLD is projected to become a prominent cause of liver transplantation.[48]

The course of the disease varies from fatty steatosis to non-alcoholic steatohepatitis (NASH).[49] 5-8% of patients who develop NASH may develop cirrhosis of the liver within five years.[50] Within three years, 12.8% of patients with liver cirrhosis develop hepatocellular carcinoma (HCC).[51] In 70% of NASH patients, metabolic syndrome and its components have NAFLD risk factors, including sedentary lifestyle and increased fat in the diet, thus considered a major cause of cryptogenic cirrhosis.[52,53]

Cardiovascular disease, chronic renal disease, insulin resistance, obesity, type 2 diabetes, postoperative difficulties after major liver surgery, and an increased risk of colorectal cancer are all linked to NAFLD.[54-56] The prevalence of NAFLD is also affected by gender, age, sleep apnea, ethnicity, and the presence of endocrine disorders (hypopituitarism, hypothyroidism, polycystic ovary syndrome, and hypogonadism).[57,58]

METABOLIC EFFECTS OF FRUCTOSE

Examining glucose metabolism will help understand the influence of fructose on fatty liver metabolism. Glucose is metabolized by glucokinase and hexokinase. Fructokinase is the enzyme that mainly breaks down fructose. Fructokinase uses adenosine triphosphate (ATP) to phosphorylate fructose to fructose-1-phosphate. Aldolase B, D-glyceraldehyde, and dihydroxyacetone phosphate are all produced. Fructose metabolism resembles glucose metabolism at this point. Glycogen and triglycerides are formed when glucose is consumed.[59] As can be seen, the first two enzymatic processes differ significantly between fructose and glucose metabolism. The liver is the primary site for fructose metabolism.[60] Fructose is different from glucose in that it can generate carbohydrate metabolites without triggering the hepatic insulin response.[61,62] In the liver, the major isoforms of fructose are ATP and intracellular phosphate.[63-66] The purine nucleotide cycle, which leads to the synthesis of uric acid, is triggered by a drop in intracellular phosphate, which activates the enzyme AMP deaminase, which converts adenosine monophosphate (AMP) to inosine monophosphate (IMP).[67] Fructose also stimulates uric acid synthesis from the amino acid precursors.[68-70] Fructose's ability to induce ATP consumption has been shown in both intravenous and human populations.[63,71] Uric acid levels rise when you consume fructose.[72,73]

Therefore, the unique thing about fructose in glucose is that it causes a temporary decrease in intracellular phosphate and ATP levels when fructose is metabolized, associated with nucleotide cycle and uric acid production. The decrease in ATP level, a disruption in protein synthesis, induction of oxidative stress, and the emergence of an important role in its fructose-mediated effects may cause its appearance by causing a number of manifestations, including mitochondrial dysfunction.[63,74] As a result of studies, the consumption of fructose has been reported to stimulate lipogenesis in animals as well and to block the oxidation of hepatic β-fatty acid.[75,76] Similarly, studies seen in humans have demonstrated that fructose intake stimulates de novo lipogenesis and prevents the oxidation of fatty acids in the liver.[76,77,78] All of these factors present fructose as sugar that causes fatty liver.

TREATMENT OPTIONS FOR FATTY LIVER

A. Treatment Methods for Alcoholic Fatty Liver Disease

The therapy of AFLD is divided into two parts: traditional treatment, new therapeutic options, and novel potential treatments

Traditional Treatment Methods:

Liver Transplantation

Liver transplantation (LT) is one of the most successful treatments for a variety of acute and chronic liver diseases, and it is one of the approaches that has lately modified the natural course of liver disease.[79,80] According to the studies, 70 nations reported 35784 liver transplants to the Global Observatory on Donation and Transplantation in 2019.[79,81]Although LT enhances the quality of life and length of life, it also carries a number of risks, including the spread of disease from the donor to the recipient.[79,82,83] Patients with severe alcoholic hepatitis related to ALD who do not react to steroids may consider LT as a last resort. Patients should abstain from alcohol for six months and receive adequate addiction therapy before undergoing liver transplantation, according to health institutes. [84] While one study suggested that a six-month abstinence period would allow the liver to heal with medical treatment and possibly not require transplantation,[85] another study noted that some improvement in liver function may occur within three months of abstinence, and many patients may die after 6 months of waiting.[86] While the three-month mortality rate is 70%, if transplantation is not performed, the mortality rate rises to 90% or more.[87,88] It is concerning because of the possibility of recurrence among patients.[89] The 10% to 50% recurrence rate after transplantation is a big issue.[90,91] However, liver transplantation due to ALD is associated with a high incidence of cardiovascular complications.[92] LT remains the only curative treatment for decompensated NAFLD cirrhosis with or without HCC.[93]

Treatment and Deprivation for Alcohol Addiction

One of the most significant processes in the treatment of ALD is limiting alcohol use, which also has an impact on death rates. The seven-year survival rate for patients who stopped alcohol was 80%, while the seven-year survival rate for those who continued to drink alcohol dropped to 50%.[94] Drugs used to treat addiction, such as naltrexone and acamprosate, have been observed to lower alcohol intake in high alcohol consumption patients,[95,96] and symptoms of topiramate withdrawal in addicts.[97]

Disulfiram, an acetaldehyde dehydrogenase inhibitor, is used to reduce alcohol intake because it causes serum acetaldehyde accumulation that causes nausea, vomiting, abdominal pain, and dizziness.[98] A γ-amino butyric acid agonist, baclofen has been found to be effective in promoting deprivation.[99] In one study, while taking 10 mg of baclofen twice a day, the daily number of beverages reduced by 68%, while when taking 20 mg of baclofen, the daily number of beverages was reduced by 53%.[100] Drugs such as naltrexone and disulfiram help maintain deprivation, and they should be avoided in patients with liver problems since they cause problems such as hepatotoxicity.[101] However, the use of acamprosate, topiramate, and baclofen in such patients is not considered objectionable.[99,102,103] Another drug used to maintain abstinence is metadoxine (MTDX). MTDX is readily absorbed and boosts acetaldehyde dehydrogenase activity when taken orally, in addition to aiding abstinence. Thus, it contributes to acute ethanol intoxication by increasing alcohol metabolism. Thus, it plays a role in acute ethanol intoxication by causing an increase in alcohol metabolism.[103] Improvement in liver function tests was observed in as little as one month after using MTDX in research of alcoholic liver patients.[104] While 74.5% of MTDX users continued their abstinence, non-users were determined to have a rate of 59.4%.[105] Most European countries utilize this medicine, which is not used in the United States.

Food and Nutritional Support

Patients with advanced ALD eat insufficiently due to several factors, including malnutrition, anorexia, and encephalopathy, with the severity often associated with this malnutrition.[94,106] Protein and energy requirements are rising in alcoholic liver patients as a result of disease stress and poor nutrition. The American College of Gastroenterology (ACG) and The American Association for the Study of Liver Diseases (AASLD) recommend 1.2 to 1.5 g/ kg of protein per day and 35 to 40 kcal/kg of calories per day for energy intake in patients with ALD.[107] The most effective way to supply this amount of calories is through enteral methods. Because enteral feeding has been shown to minimize complications and improve one-year mortality in patients in important research.[108,109] In patients in coma, parenteral nutritional support may be necessary. The amounts of vitamins (folate, vitamin B6, vitamin B12,[110,111] vitamin A, and thiamine[112]) and minerals (selenium, zinc, copper, and magnesium) in ALD are thought to vary, and these variations may have an impact on liver disease.[113] It has been observed that zinc levels decrease especially in ALD patients and animal models, and it has been shown that ALD improvement occurs as a result of supplementation of this mineral.[114]

Corticosteroids and Glucocorticosteroids

Infection is one of the most common causes of death in patients with alcoholic hepatitis. Factors such as malnutrition and medical procedures increase the risk of infection. Hepatitis C virus (HCV) promotes liver damage and the development of HCC, making it a risk factor for ALD progression.[115,116] This effect occurs when both alcohol and HCV cause a change in cellular immunity, increasing free radical oxidative damage and causing the HCV to self-replicate if exposed to alcohol. Therefore, the occurrence of an infection along with the disease reduces the chance of survival.[117,118] Alcoholic patients with HCV infection have a 30-fold increased risk of cirrhosis and a two to eight-fold increased risk of death when compared to those who do not have HCV infection.[119,120] Based on this information, HCV screening should be screened for all ALD patients before treatment begins and all HCV patients should be advised to restrict their alcohol consumption.[121]

Corticosteroids have potent anti-inflammatory properties and are useful in the treatment of autoimmune hepatitis. To date, many different clinical investigations on the use of corticosteroids to treat individuals with ALD have been undertaken.[122-124] While it can be said that the use of corticosteroids is beneficial, although not valid for all patients, 40% of the patients do not respond to corticosteroids.[125] In a meta-analysis combining data from 3 randomized control studies, it was found that when suitable patients were treated with 40 mg/day prednisolone (a synthetic corticosteroid) for 28 days, the survival rate in the placebo was 85%, compared with 65 % for patients on glucocorticosteroid, while the death rate dropped from 35% to 15% for steroids.[124] Although some patients have a beneficial effect by responding partially to steroid treatment, it has been observed that patients have not responded. It is recommended to stop steroid therapy in patients who do not respond.[126] Steroids are usually avoided in patients with active infection, gastrointestinal bleeding, chronic hepatitis B virus infection, or hepatorenal syndrome (HRS).[127] Therefore, such patients can be treated with pentoxifylline (PTX) as a second line.[128]

New Therapeutic Options and Novel Potential Treatments:

S-Adenosylmethionine

S-adenosylmethionine (SAM) is a methyl donor involved in many methylation reactions critical to normal liver function. SAM levels have been reported to be below in ALD patients. Therefore, raising SAM levels is thought to be a potential therapy. Various animal studies have seen that preventing a decline in SAM levels can prevent liver injury.[129]

Various Chemokines and Interleukins

Chemokines play a key role in the pathogenesis of alcoholic hepatitis. Many distinct chemokines, such as CXCL5, CXCL6, CXCL10, and CCL20, have been found to be highly elevated in the ASH liver compared to normal liver levels in studies.[130,131] Interleukin-8 (IL-8) is one of the most prominent neutrophil chemoattractants, and high IL-8 levels are associated with the severity of alcoholic hepatitis.[130] IL-22, a member of the interleukin-10 (IL-10) family that inhibits the production of proinflammatory cytokines, is implicated in bacterial infections and tissue repair.[132] IL-22 contains anti-steatotic and antioxidant properties, and it could be used to treat ALD patients. Also, auxiliary T cell levels involved in IL-22 production are found to be associated with improvement in alcoholic hepatitis patients.[133] In ethanol-fed mice, administration of recombinant IL-22 has improved the condition,[134] whereas blocking the IL-22 receptor has worsened the condition.[135]

Endocannabinoids Antagonists

In experiments on animal models of alcoholic liver injury, researchers are determined that mice lacking cannabinoid receptor type 1 (CB1) are resistant to fatty liver damage, but mice lacking cannabinoid receptor type 2 (CB2) are more susceptible.[136,137]

Osteopontin Inhibition

Osteopontin (OPN), an extracellular matrix protein, is involved in wound healing in response to injury in many organs.[138] Another study observed that mice without OPN had weakened due to alcohol-induced liver disease.[139]

B. Treatments of Non-Alcoholic Fatty Liver Disease

Although NAFLD treatment methods have common points with AFLD treatment methods, they differ in some ways.

Lifestyle Change

A lifestyle change aimed at increasing weight loss and physical activity is critical for those with NAFLD. It is possible that patients will be advised to lose 10% or more of their body weight. This state is associated with an improved cardiovascular risk profile and steatosis in the patients.[140] Hepatic inflammation and a reduction in hepatocellular damage can be seen even with a 7-9% weight loss.[141,142] In studies, patients who got dietary treatment and engaged in moderate physical activity for 200 minutes per week for 48 weeks had less steatosis and inflammation in their liver biopsy, as well as a decrease in body weight.[141]

Diet

Patients should follow a calorie-restricted diet targeted at losing 0.5-1 kg/week until reaching their target weight.[143] It is suggested that as the diet advice avoid saturated fats, simple carbohydrates, and sugary drinks.[144,145] Recently, there has been an increase in interest in omega-3 (ω-3) polyunsaturated fatty acids (n-3 PUFAs) in the diet. Studies show that NAFLD patients consume less n-3 PUFA.[146,147] In studies a reduction in liver fat with n-3 PUFA supplementation was, but no significant reduction in ALT (Alanine aminotransferase) levels.[148] Adding fish oil to the diet may offer treatment options for patients. Dietitian intervention is also important for these patients. Lifestyle changes in dietitian control may be preferred in terms of observing improvement in weight loss NAFLD.[149]

Exercise

Aerobic exercise improves skeletal muscle insulin sensitivity and lowers insulin resistance, which is one of the important mechanisms that lead to NAFLD.[150,151] Studies examining moderate-violence, high-violence, and resistance exercise have associated improvements in liver enzymes and a reduction in steatosis, independent of weight loss.[152-154] Individuals with NAFLD should be advised to increase physical activity. There are some approaches that propose 30 minutes of moderate exercise five times a week.[155] It has been observed that individuals with NAFLD are less active than healthy individuals.[156,157] There are studies showing that they do not exercise much and are less ready for lifestyle changes.[158,159]

Bariatric Surgery

Bariatric surgery is becoming important in the treatment of obese patients with NAFLD. Weight loss from bariatric surgery increases insulin sensitivity and has specific effects on liver histology.[160] However, it is not recommended as first-line therapy because of the lack of long-term effect data and the risk of postoperative hepatic failure.[161,162] In adults with a body mass index (BMI) > 50, surgery may be considered the first treatment.[143]

Vitamin E

Experiments with vitamin E have been linked to improved liver function and lower levels of oxidative stress indicators. However, improvement in the disease's histology grading is less substantial.[163,164] In another study, it was observed that vitamin E was associated with a decrease in steatohepatitis in the childhood NASH study.[165] A study observed that people who took vitamins E and C for six months were no better than those who took a placebo in treating NASH.[166] Six-month research comparing the combination of pioglitazone and vitamin E to vitamin E alone observed that both groups had lower serum ALT levels, but the combination group had a significant histological improvement.[167] According to meta-analysis research, high-dose vitamin E supplementation (>400 IU/day) is linked to an increase in all-cause mortality and cardiovascular fatalities, reversing the trend toward vitamin E therapy.[168]

Pentoxifylline

Pentoxifylline inhibits proinflammatory cytokines, including TNF-α. In vitro studies on hepatic stellate cells (HSC) are observed anti-fibrogenic effects.[169,170] Following a one-year treatment with PTX, researchers found two percentage point reduction in NAFLD activity in patients. In addition, steatosis, inflammation, and fibrosis were also produced significant improvements using PTX.[171] However, a randomized, multicenter double-blind research undertaken in 65 institutions in the United Kingdom with over 1,000 patients indicated that PTX had no effect on survival or disease progression in patients with severe NASH.[172,173]

Metformin

Metformin is preferred in the first stage because it improves insulin sensitivity and is an agent for type 2 diabetes mellitus (T2DM). Widely tested in NAFLD. This is because insulin resistance is an important pathogenic feature of T2DM patients. Studies with metformin have shown that there may be a histological benefit.[174,175] However, other randomized controlled studies have yielded negative results.[176,177] In other meta-analysis studies, it was also found that Metformin had no therapeutic effects on liver histology in NASH and NAFLD patients.[178,179]

Angiotensin Receptor Blockers

Non-alcoholic fatty liver disease is associated with metabolic syndrome and hypertension is a major component of metabolic syndrome. Therefore, Angiotensin receptor blockers may be part of the NAFLD treatment. A study in NAFLD patients was observed that losartan (angiotensin II receptor antagonist) treatment was associated with improvement in necro-inflammation and fibrosis.[180]

Agents to Lower Lipids

Studies have shown that gemfibrozil is associated with an improvement in lower levels in NAFLD patients compared to placebo.[181] Clofibrate, on the other hand, had no beneficial effect on liver tests or histology scores.[182] In investigations, ezetimibe (an inhibitor of intestinal lipid reabsorption) decreased blood TNF-α, hepatic lipid content, and ALT levels in NAFLD mice.[183,184]

Ursodeoxycholic Acid

Ursodeoxycholic acid (UDCA) is a naturally-occurring bile acid commonly used mostly for chronic cholestatic liver disease.[185] UDCA reduces oxidative stress. It has been explained by studies that it has various anti-apoptotic, antioxidant, and anti-inflammatory properties.[186-189] In studies are observed that UDCA improves liver enzymes and hepatic steatosis in patients with NAFLD.[190,191]

Anti-Obesity Drugs

Non-alcoholic fatty liver disease is exacerbated by obesity. Therefore, anti-obesity medications and pharmacological agents can be evaluated as viable candidates for controlling and treating the disease process. However, data on the efficiency of anti-obesity medicines other than orlistat in the treatment of NAFLD are scarce.[192]

Orlistat

Orlistat is an anti-obesity drug that inhibits fat absorption. A study showed an improving effect on liver enzymes in NAFLD patients but had no effect on liver fibrosis score.[193] In another study, the effect of orlistat on NAFLD was evaluated in patients who received a 1400 kcal diet and vitamin E (800 IU) per day. Orlistat (120 mg three times daily) did not increase weight loss or improve liver enzymes and histopathology in 50 overweight subjects when given.[194]

Lorcaserin

Lorcaserin is a serotonin 2C receptor agonist that improves T2DM and promotes weight loss. In a six-month, randomized, placebo-controlled double-blind study, lorcaserin reduced the fatty liver index in 48 patients without T2DM.[195]

Probiotics

Intestinal bacterial overgrowth increased intestinal permeability, and increased paracellular leakage of intestinal luminal antigens are all variables that promote the development of NASH in patients with NAFLD, just as they are in individuals with AFLD. As a way, probiotics could be a viable treatment option for NASH patients.[196,197] There has been improvement in liver enzymes relative to placebo in NAFLD patients who were treated in a randomized controlled trial using Streptococcus thermophilus and Lactobacillus bulgaricus.[198] In another randomization study, a significant reduction in steatosis, TNF-α, AST, and NASH was observed in the combination treatment group, with patients receiving Bifidobacterium longum in combination with fructooligosaccharides, within the lifestyle modification group alone, and lifestyle modification (exercise and diet).[199]

Liver Transplantation

Since the increasing prevalence of NAFLD, liver transplantation is an increasing treatment modality for NASH cirrhosis.[200] In a study in which liver transplantation was performed in 98 patients for NASH cirrhosis, recurring steatosis was observed in 70% of the patients, and NASH was observed in 25% of the patients. However, none of the patients developed graft failure or required retransplantation[201] for three years.[202]

In conclusion, the liver is the body's largest, most important organ, and is involved in a variety of physiological functions. Fatty liver is examined as alcoholic and non-alcoholic. Alcoholic liver steatosis is associated with increased consumption of alcohol, while it is not possible to show a single reason for non-alcoholic fatty liver disease. NAFLD is also referred to as MAFLD. The causes that may cause the disease can be many diseases such as obesity, sedentary life, hyperlipidemia, insulin resistance, T2DM. The prevalence of fatty liver is influenced by gender, age, sleep apnea, ethnicity, endocrine disorders. The course of the disease varies from cirrhosis to HCC. Causes of this disease are being investigated, more and more the efforts to treat it are increasing and the studies on the fatty liver are of interest. Many studies have been conducted on fatty and NAFLD. However, no single and definitive treatment is available. Different treatments are preferred depending on many factors such as the characteristics of the patient, the degree of disease, and the cause of fatty liver. Nowadays, new methods are being researched and applied apart from the treatment methods from the past. Combining old and modern therapy strategies appears to be a viable option for treating ALD and NAFLD. In order to ensure trust and prove the effectiveness of these treatments, additional research should be undertaken on the treatments that will be acknowledged as the new therapy approach. In this way, the most appropriate treatment methods can be found and applied to patients.

Cite this article as: Topal E, Aydemir K, Çağlar Ö, Arda B, Kayabaşı O, Yıldız M, Özyılmaz E, Erbaş O. Fatty Liver Disease: Diagnosis and Treatment. JEB Med Sci 2021;2(3):343-357.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

References

  1. Çağlar O, Özyılmaz E. Effect of Quercetin in Hepatocellular Carcinoma. Research & Reviews in Health Sciences, 2020;311-28.
  2. Michalopoulos GK, Bhushan B. Liver regeneration: biological and pathological mechanisms and implications. Nat Rev Gastroenterol Hepatol. 2021;18:40-55.
  3. Bademci R, Erdoğan MA, Eroğlu E, Meral A, Erdoğan A, Atasoy Ö, et al. Demonstration of the protective effect of ghrelin in the livers of rats with cisplatin toxicity. Hum Exp Toxicol. 2021;40:2178-87.
  4. Highton AJ, Schuster IS, Degli-Esposti MA, Altfeld M. The role of natural killer cells in liver inflammation. Semin Immunopathol. 2021;43:519-33.
  5. Trefts E, Gannon M, Wasserman DH. The liver. Curr Biol. 2017 6;27:R1147-R51.
  6. Güzel S, Şahinoğulları ZU, Canacankatan N, Antmen ŞE, Kibar D, Bayrak G. The ameliorating effect of silymarin against vancomycin-induced apoptosis and inflammation in rat liver. J Res Pharm, 2019;23, 719-28.
  7. Wong VW, Wong GL, Woo J, Abrigo JM, Chan CK, Shu SS, et al. Impact of the New Definition of Metabolic Associated Fatty Liver Disease on the Epidemiology of the Disease. Clin Gastroenterol Hepatol. 2021;19:2161-71.e5.
  8. Yilmaz Y, Yilmaz N, Ates F, Karakaya F, Gokcan H, Kaya E, et al. The prevalence of metabolic-associated fatty liver disease in the Turkish population: A multicenter study. In Hepatology Forum 2021;2: 37.
  9. Makri E, Goulas A, Polyzos SA. Epidemiology, pathogenesis, diagnosis and emerging treatment of nonalcoholic fatty liver disease. Archives of medical research, 2021;52, 25-37.
  10. Aron-Wisnewsky J, Vigliotti C, Witjes J, Le P, Holleboom AG, Verheij J, et al. Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol. 2020; 17:279-97.
  11. Kaya E, Yılmaz Y. Non-alcoholic fatty liver disease: A growing public health problem in Turkey. Turk J Gastroenterol. 2019;30:865-71.
  12. Murag S, Ahmed A, Kim D. Recent Epidemiology of Nonalcoholic Fatty Liver Disease. Gut Liver. 2021;15:206-16.
  13. Freitas-Lopes MA, Mafra K, David BA, Carvalho-Gontijo R, Menezes GB. Differential Location and Distribution of Hepatic Immune Cells. Cells. 2017;7;6:48.
  14. Tanimine N, Tanaka Y, Abe T, Piao J, Chayama K, Ohdan H. Functional Behavior of NKp46-Positive Intrahepatic Natural Killer Cells Against Hepatitis C Virus Reinfection After Liver Transplantation. Transplantation. 2016;100:355-64.
  15. Koyama Y, Brenner DA. Liver inflammation and fibrosis. J Clin Invest. 2017;127:55-64.
  16. Solmaz V, Tekatas A, Erdoğan MA, Erbaş O. Exenatide, a GLP-1 analog, has healing effects on LPS-induced autism model: Inflammation, oxidative stress, gliosis, cerebral GABA, and serotonin interactions. Int J Dev Neurosci. 2020;80:601-12.
  17. Mountford S, Effenberger M, Noll-Puchta H, Griessmair L, Ringleb A, Haas S, et al. Modulation of Liver Inflammation and Fibrosis by Interleukin-37. Front Immunol. 2021;12:603649.
  18. Ekmekçi AM, Balandi F, Erbaş O. RIPK1 and obesity-induced inflammation. D J Tx Sci 2021;6:17-22
  19. Ju L, Han J, Zhang X, Deng Y, Yan H, Wang C, et al. Obesity-associated inflammation triggers an autophagy-lysosomal response in adipocytes and causes degradation of perilipin 1. Cell Death Dis 2019;10:121.
  20. Scott A, Khan KM, Cook JL, Duronio V. What is “inflammation”? Are we ready to move beyond Celsus?. British journal of sports medicine 2004;38, 248-9.
  21. Meng H, Niu R, You H, Wang L, Feng R, Huang C, et al. Interleukin-9 attenuates inflammatory response and hepatocyte apoptosis in alcoholic liver injury. Life Sci. 2022;288:120180.
  22. Erbaş O, Solmaz V, Aksoy D, Yavaşoğlu A, Sağcan M, Taşkıran D. Cholecalciferol (vitamin D 3) improves cognitive dysfunction and reduces inflammation in a rat fatty liver model of metabolic syndrome. Life Sci. 2014;103:68-72.
  23. Aksoy D, Solmaz V, Taşkıran D, Erbaş O. The association between seizure predisposition and inflammation in a rat model of fatty liver disease. Neurol Sci. 2014;35:1441-6.
  24. Gao B., Tsukamoto H. Inflammation in alcoholic and nonalcoholic fatty liver disease: friend or foe?. Gastroenterology 2016;150, 1704-9.
  25. Lazaro R, Wu R, Lee S, Zhu NL, Chen CL, French SW, et al. Osteopontin deficiency does not prevent but promotes alcoholic neutrophilic hepatitis in mice. Hepatology. 2015; 61:129-40.
  26. Braunersreuther V, Viviani GL, Mach F, Montecucco F. Role of cytokines and chemokines in non-alcoholic fatty liver disease. World J Gastroenterol. 2012;18:727-35.
  27. Erbaş O, Akseki HS, Solmaz V, Aktuğ H, Taşkıran D. Fatty liver-induced changes in stereotypic behavior in rats and effects of glucagon-like peptide-1 analog on stereotypy. The Kaohsiung J Med Sci 2014; 30, 447-52.
  28. Hamada K, Wang P, Xia Y, Yan N, Takahashi S, Krausz KW, et al. Withaferin A alleviates ethanol-induced liver injury by inhibiting hepatic lipogenesis. Food Chem Toxicol. 2022;112807.
  29. Osna NA, Donohue TM Jr, Kharbanda KK. Alcoholic Liver Disease: Pathogenesis and Current Management. Alcohol Res. 2017;38:147-61.
  30. Yenigün M. Alcohol Consumption and Medicine. Med Bull Haseki 2006;44.
  31. Vizuete AFK, Mussulini BH, Zenki KC, Baggio S, Pasqualotto A, Rosemberg DB, et al. Prolonged ethanol exposure alters glutamate uptake leading to astrogliosis and neuroinflammation in adult zebrafish brain. Neurotoxicol. 2022;88:57-64.
  32. Finelli R, Mottola F, Agarwal A. Impact of Alcohol Consumption on Male Fertility Potential: A Narrative Review. Int J Environ Res Public Health.2022;19, 328.
  33. Cederbaum AI. Alcohol metabolism. Clin Liver Dis. 2012;16, 667-85.
  34. Lu Y, Cederbaum AI. CYP2E1 and oxidative liver injury by alcohol. Free Radical Biology and Medicine 2008;44, 723-38.
  35. Rocco A, Compare D, Angrisani D, Sanduzzi Zamparelli M, Nardone G. Alcoholic disease: liver and beyond. World J Gastroenterol. 2014;20:14652-9.
  36. Edenberg HJ. The genetics of alcohol metabolism: role of alcohol dehydrogenase and aldehyde dehydrogenase variants. Alcohol Res Health. 2007;30:5-13.
  37. Schuckit M.A. Ethanol and Methanol. In: Brunton L.L, ed. Goodman & Gilman’s, The Pharmacological Basis of Therapeutics. 12nd ed. New York: McGraw-Hill; 2011, p: 629-47.
  38. Dawson DA, Grant BF, Li TK. Quantifying the risks associated with exceeding recommended drinking limits. Alcohol Clin Exp Res 2005;29:902-8.
  39. NIAAA. Alcohol Facts and Statistics. 2021. Available from: https://www.niaaa.nih.gov/publications/brochures-andfact-sheets/alcohol-facts-and-statistics
  40. Lane BP, Lieber CS. Ultrastructural alterations in human hepatocytes following ingestion of ethanol with adequate diets. Am J Pathol 1966;49:593-603.
  41. Kaya E, Yilmaz Y. Metabolic-associated Fatty Liver Disease (MAFLD): A Multi-systemic Disease Beyond the Liver. J Clin Transl Hepatol. 2021; 0-0.
  42. Eren F, Kaya E, Yilmaz Y. Accuracy of Fibrosis-4 index and non-alcoholic fatty liver disease fibrosis scores in metabolic (dysfunction) associated fatty liver disease according to body mass index: failure in the prediction of advanced fibrosis in lean and morbidly obese individuals. Eur J Gastroenterol Hepatol. 2022;34:98-103.
  43. Eslam M, Sanyal AJ, George J; International Consensus Panel. MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology 2020;158:1999-2014.e1.
  44. Younossi Z, Tacke F, Arrese M, Chander Sharma B, Mostafa I, Bugianesi E, et al. Global Perspectives on Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Hepatology. 2019;69:2672-82.
  45. Fatima K, Moeed A, Waqar E, Atif AR, Kamran A, Rizvi H, et al. Efficacy of statins in treatment and development of non-alcoholic fatty liver disease and steatohepatitis: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol. 2021;46:101816.
  46. Hojland Ipsen D, Tveden-Nyborg P, Lykkesfeldt J. Normal weight dyslipidemia: is it all about the liver? Obesity (Silver Spring) 2016;24:556-67.
  47. Wattacheril J, Sanyal AJ. Lean NAFLD: an underrecognized outlier. Curr Hepatol Rep. 2016;15:134-9.
  48. Wong RJ, Aguilar M, Cheung R, Perumpail RB, Harrison SA, Younossi ZM, et al. Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Gastroenterology. 2015;148:547-55.
  49. Sanyal AJ, Brunt EM, Kleiner DE, Kowdley KV, Chalasani N, Lavine JE, et al. Endpoints and clinical trial design for nonalcoholic steatohepatitis. Hepatol. 2011;54:344-53.
  50. Ekstedt M, Franzén LE, Mathiesen UL, Thorelius L, Holmqvist M, Bodemar G, et al. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatol. 2006;44:865-73.
  51. White DL, Kanwal F, El-Serag HB. Association between nonalcoholic fatty liver disease and risk for hepatocellular cancer, based on systematic review. Clin Gastroenterol Hepatol. 2012;10:1342-1359.e2.
  52. Ortiz-Lopez C, Lomonaco R, Orsak B, Finch J, Chang Z, Kochunov VG, et al. Prevalence of prediabetes and diabetes and metabolic profile of patients with nonalcoholic fatty liver disease (NAFLD) Diabetes Care. 2012;35:873-8.
  53. Souza MR, Diniz Mde F, Medeiros-Filho JE, Araújo MS. Metabolic syndrome and risk factors for non-alcoholic fatty liver disease. Arq Gastroenterol. 2012;49:89-96.
  54. Anstee QM, Targher G, Day CP. Progression of NAFLD to diabetes mellitus, cardiovascular disease or cirrhosis. Nat Rev Gastroenterol Hepatol. 2013;10:330-44.
  55. Lonardo A, Sookoian S, Chonchol M, Loria P, Targher G. Cardiovascular and systemic risk in nonalcoholic fatty liver disease - atherosclerosis as a major player in the natural course of NAFLD. Curr Pharm Des. 2013;19:5177-92.
  56. Vanni E, Bugianesi E, Kotronen A, De Minicis S, Yki-Järvinen H, Svegliati-Baroni G. From the metabolic syndrome to NAFLD or vice versa? Dig Liver Dis. 2010;42:320-30.
  57. Loria P, Carulli L, Bertolotti M, Lonardo A. Endocrine and liver interaction: the role of endocrine pathways in NASH. Nat Rev Gastroenterol Hepatol. 2009;6:236-47.
  58. Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther. 2011;34:274-85.
  59. Steinmann B, Gitzelmann R, Van den Berghe G. Disorders of Fructose Metabolism. In: Scriver C, Beaudet A, Sly W, Valle D, editors. The Metabolic and Molecular Basis of Inherited Disease. New York: McGraw-Hill; 2001. pp. 1489-520.
  60. Mayes PA. Intermediary metabolism of fructose. Am. J. Clin. Nutr. 1993;58:754s-765s.
  61. Li MV, Chen W, Harmancey RN, Nuotio-Antar AM, Imamura M, Saha P, et al. Glucose-6-phosphate mediates activation of the carbohydrate responsive binding protein (ChREBP) Biochem. Biophys. Res. Commun. 2010;395:395-400.
  62. Arden C, Tudhope SJ, Petrie JL, Al-Oanzi ZH, Cullen KS, Lange AJ, et al. Fructose-2,6-bisphosphate is essential for glucose-regulated gene transcription of glucose6-phosphatase and other ChREBP target genes in hepatocytes. Biochem. J. 2012;443:111-123.
  63. Maenpaa PH, Raivio KO, Kekomaki MP. Liver adenine nucleotides: fructose-induced depletion and its effect on protein synthesis. Science. 1968;161:1253-4.
  64. Smith CM, Rovamo LM, Raivio KO. Fructose-induced adenine nucleotide catabolism in isolated rat hepatocytes. Can J Biochem. 1977;55:1237-40.
  65. van den Berghe G, Bronfman M, Vanneste R, Hers HG. The mechanism of adenosine triphosphate depletion in the liver after a load of fructose. A kinetic study of liver adenylate deaminase. Biochem J. 1977;162:601-9.
  66. Kurtz TW, Kabra PM, Booth BE, Al-Bander HA, Portale AA, Serena BG, et al. Liquid-chromatographic measurements of inosine, hypoxanthine, and xanthine in studies of fructose-induced degradation of adenine nucleotides in humans and rats. Clin Chem. 1986;32:782-6.
  67. Van den Berghe G. Fructose: metabolism and short-term effects on carbohydrate and purine metabolic pathways. Prog Biochem Pharmacol. 1986;21:1-32.
  68. Emmerson BT. Effect of oral fructose on urate production. Ann Rheum Dis. 1974;33:276-80.
  69. Raivio KO, Becker A, Meyer LJ, Greene ML, Nuki G, Seegmiller JE. Stimulation of human purine synthesis de novo by fructose infusion. Metabolism. 1975;24:861-9.
  70. Jensen T, Abdelmalek MF, Sullivan S, Nadeau KJ, Green M, Roncal C, et al. Fructose and sugar: A major mediator of non-alcoholic fatty liver disease. J Hepatol. 2018;68:1063-1075.
  71. Abdelmalek MF, Lazo M, Horska A, Bonekamp S, Lipkin EW, Balasubramanyam A, et al. Higher dietary fructose is associated with impaired hepatic adenosine triphosphate homeostasis in obese individuals with type 2 diabetes. Hepatol. 2012;56:952-60.
  72. Perheentupa J, Raivio K. Fructose-induced hyperuricaemia. Lancet. 1967;2:528-31.
  73. Stirpe F, Della Corte E, Bonetti E, Abbondanza A, Abbati A, De Stefano F. Fructose-induced hyperuricaemia. Lancet. 1970;2:1310-1.
  74. Choi YJ, Shin HS, Choi HS, Park JW, Jo I, Oh ES, et al. Uric acid induces fat accumulation via generation of endoplasmic reticulum stress and SREBP-1c activation in hepatocytes. Lab Invest. 2014;94:1114-25.
  75. Lanaspa MA, Sanchez-Lozada LG, Choi YJ, Cicerchi C, Kanbay M, Roncal-Jimenez CA, et al. Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: potential role in fructose-dependent and -independent fatty liver. J Biol Chem. 2012;287:40732-44.
  76. Softic S, Cohen DE, Kahn CR. Role of Dietary Fructose and Hepatic De Novo Lipogenesis in Fatty Liver Disease. Dig Dis Sci. 2016;61:1282-93.
  77. Faeh D, Minehira K, Schwarz JM, Periasamy R, Park S, Tappy L. Effect of fructose overfeeding and fish oil administration on hepatic de novo lipogenesis and insulin sensitivity in healthy men. Diabetes. 2005;54:1907-13.
  78. Stanhope KL, Schwarz JM, Keim NL, Griffen SC, Bremer AA, Graham JL, et al. Consuming fructose-sweetened, not glucose-sweetened, beverages increases visceral adiposity and lipids and decreases insulin sensitivity in overweight/obese humans. J Clin Invest. 2009;119:1322-34.
  79. Domínguez-Gil B, Moench K, Watson C, Serrano MT, Hibi T, Asencio JM, et al. Prevention and Management of Donor-transmitted Cancer After Liver Transplantation: Guidelines From the ILTS-SETH Consensus Conference. Transplantation. 2022;106:e12-e29.
  80. Taşkıran E, Akar H, Yıldırım M, Erbaş O. Liver transplantation: indications, contraindications, rejection and longterm follow-up. FNG & Bilim Tıp Transplantasyon Dergisi, 2016;1:59-66.
  81. Global Observatory on Donation and Transplantation. WHO-ONT. Available at http://www.transplantobservatory.org. Accessed June 2021.
  82. Merion RM, Schaubel DE, Dykstra DM, Freeman RB, Port FK, Wolfe RA. The survival benefit of liver transplantation. Am J Transplant. 2005;5:307-13.
  83. Yang LS, Shan LL, Saxena A, Morris DL. Liver transplantation: a systematic review of long-term quality of life. Liver Int. 2014;34:1298-313.
  84. Testino G, Burra P, Bonino F, Piani F, Sumberaz A, Peressutti R, et al. Acute alcoholic hepatitis, end stage alcoholic liver disease and liver transplantation: an Italian position statement. World J Gastroenterol. 2014;20: 14642-14651.
  85. Lucey MR, Brown KA, Everson GT, Fung JJ, Gish R, Keeffe EB, et al. Minimal criteria for placement of adults on the liver transplant waiting list: a report of a national conference organized by the American Society of Transplant Physicians and the American Association for the Study of Liver Diseases. Liver Transpl Surg. 1997;3: 628-637.
  86. Veldt BJ, Lainé F, Guillygomarc’h A, Lauvin L, Boudjema K, Messner M, et al. Indication of liver transplantation in severe alcoholic liver cirrhosis: quantitative evaluation and optimal timing. J Hepatol. 2002;36: 93-98.
  87. Testino G, Sumberaz A, Borro P. Comment to “liver transplantation for patients with alcoholic liver disease: an open question.” Digestive and liver disease: official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver. 2013; 80-81.
  88. Testino G, Ferro C, Sumberaz A, Messa P, Morelli N, Guadagni B, et al. Type-2 hepatorenal syndrome and refractory ascites: role of transjugular intrahepatic portosystemic stent-shunt in eighteen patients with advanced cirrhosis awaiting orthotopic liver transplantation. Hepatogastroenterology. 2003;50: 1753-1755.
  89. Tan H-H, Virmani S, Martin P. Controversies in the management of alcoholic liver disease. Mt Sinai J Med. 2009;76: 484-498.
  90. Burra P, Mioni D, Cecchetto A, Cillo U, Zanus G, Fagiuoli S, et al. Histological features after liver transplantation in alcoholic cirrhotics. J Hepatol. 2001;34: 716-722.
  91. Pageaux G-P, Bismuth M, Perney P, Costes V, Jaber S, Possoz P, et al. Alcohol relapse after liver transplantation for alcoholic liver disease: does it matter? J Hepatol. 2003;38: 629-634.
  92. Burra P, Senzolo M, Adam R, Delvart V, Karam V, Germani G, et al. Liver transplantation for alcoholic liver disease in Europe: a study from the ELTR (European Liver Transplant Registry). Am J Transplant. 2010;10: 138-148.
  93. Villeret F, Dumortier J, Erard-Poinsot D. How will NAFLD change the liver transplant landscape in the 2020s? Clin Res Hepatol Gastroenterol. 2021;46:101759.
  94. Alexander JF, Lischner MW, Galambos JT. Natural history of alcoholic hepatitis. II. The long-term prognosis. Am J Gastroenterol. 1971;56: 515-525.
  95. Roozen HG, de Waart R, van der Windt DAWM, van den Brink W, de Jong CAJ, Kerkhof AJFM. A systematic review of the effectiveness of naltrexone in the maintenance treatment of opioid and alcohol dependence. Eur Neuropsychopharmacol. 2006;16: 311-323.
  96. Mason BJ, Lehert P. Acamprosate for alcohol dependence: a sex-specific meta-analysis based on individual patient data. Alcohol Clin Exp Res. 2012;36: 497-508.
  97. Kenna GA, Lomastro TL, Schiesl A, Leggio L, Swift RM. Review of topiramate: an antiepileptic for the treatment of alcohol dependence. Curr Drug Abuse Rev. 2009;2: 135-142.
  98. Fuller RK, Branchey L, Brightwell DR, Derman RM, Emrick CD, Iber FL, et al. Disulfiram treatment of alcoholism. A Veterans Administration cooperative study. JAMA. 1986;256: 1449-1455.
  99. Addolorato G, Leggio L, Ferrulli A, Cardone S, Vonghia L, Mirijello A, et al. Effectiveness and safety of baclofen for maintenance of alcohol abstinence in alcohol-dependent patients with liver cirrhosis: randomised, double-blind controlled study. Lancet. 2007;370: 1915-1922.
  100. Addolorato G, Leggio L, Ferrulli A, Cardone S, Bedogni G, Caputo F, et al. Dose-response effect of baclofen in reducing daily alcohol intake in alcohol dependence: secondary analysis of a randomized, double-blind, placebo-controlled trial. Alcohol Alcohol. 2011;46: 312-317.
  101. Addolorato G, Russell M, Albano E, Haber PS, Wands JR, Leggio L. Understanding and treating patients with alcoholic cirrhosis: an update. Alcohol Clin Exp Res. 2009;33: 1136-1144.
  102. Haass-Koffler CL, Leggio L, Kenna GA. Pharmacological approaches to reducing craving in patients with alcohol use disorders. CNS Drugs. 2014;28: 343-360.
  103. Vuittonet CL, Halse M, Leggio L, Fricchione SB, Brickley M, Haass-Koffler CL, et al. Pharmacotherapy for alcoholic patients with alcoholic liver disease. Am J Health Syst Pharm. 2014;71: 1265-1276.
  104. Caballería J, Parés A, Brú C, Mercader J, García Plaza A, Caballería L, et al. Metadoxine accelerates fatty liver recovery in alcoholic patients: results of a randomized double-blind, placebo-control trial. Spanish Group for the Study of Alcoholic Fatty Liver. J Hepatol. 1998;28: 54-60.
  105. Higuera-de la Tijera F, Servín-Caamaño AI, Serralde-Zúñiga AE, Cruz-Herrera J, Pérez-Torres E, Abdo-Francis JM, et al. Metadoxine improves the threeand six-month survival rates in patients with severe alcoholic hepatitis. World J Gastroenterol. 2015;21: 4975-4985.
  106. Mendenhall C, Roselle GA, Gartside P, Moritz T. Relationship of protein calorie malnutrition to alcoholic liver disease: a reexamination of data from two Veterans Administration Cooperative Studies. Alcohol Clin Exp Res. 1995;19: 635-641.
  107. Plauth M, Cabré E, Riggio O, Assis-Camilo M, Pirlich M, Kondrup J, et al. ESPEN Guidelines on Enteral Nutrition: Liver disease. Clin Nutr. 2006;25: 285-294.
  108. Henkel AS, Buchman AL. Nutritional support in patients with chronic liver disease. Nat Clin Pract Gastroenterol Hepatol. 2006;3: 202-209.
  109. Cabré E, Rodríguez-Iglesias P, Caballería J, Quer JC, Sánchez-Lombraña JL, Parés A, et al. Short- and longterm outcome of severe alcohol-induced hepatitis treated with steroids or enteral nutrition: a multicenter randomized trial. Hepatology. 2000;32: 36-42.
  110. Fragasso A, Mannarella C, Ciancio A, Scarciolla O, Nuzzolese N, Clemente R, et al. Holotranscobalamin is a useful marker of vitamin B12 deficiency in alcoholics. ScientificWorldJournal. 2012;2012: 128182.
  111. Rossi RE, Conte D, Massironi S. Diagnosis and treatment of nutritional deficiencies in alcoholic liver disease: Overview of available evidence and open issues. Dig Liver Dis. 2015;47: 819-825.
  112. McClain CJ, Barve SS, Barve A, Marsano L. Alcoholic liver disease and malnutrition. Alcohol Clin Exp Res. 2011;35: 815-820.
  113. Halsted CH. Nutrition and alcoholic liver disease. Semin Liver Dis. 2004;24: 289-304.
  114. Kang YJ, Zhou Z. Zinc prevention and treatment of alcoholic liver disease. Mol Aspects Med. 2005;26: 391-404.
  115. Donato F, Tagger A, Chiesa R, Ribero ML, Tomasoni V, Fasola M, et al. Hepatitis B and C virus infection, alcohol drinking, and hepatocellular carcinoma: a case-control study in Italy. Brescia HCC Study. Hepatology. 1997;26: 579-584.
  116. Hutchinson SJ, Bird SM, Goldberg DJ. Influence of alcohol on the progression of hepatitis C virus infection: a meta-analysis. Clin Gastroenterol Hepatol. 2005;3: 1150-1159.
  117. Befrits R, Hedman M, Blomquist L, Allander T, Grillner L, Kinnman N, et al. Chronic hepatitis C in alcoholic patients: prevalence, genotypes, and correlation to liver disease. Scand J Gastroenterol. 1995;30: 1113-1118.
  118. Chen CM, Yoon Y-H, Yi H-Y, Lucas DL. Alcohol and hepatitis C mortality among males and females in the United States: a life table analysis. Alcohol Clin Exp Res. 2007;31: 285-292.
  119. Henry JA, Moloney C, Rivas C, Goldin RD. Increase in alcohol related deaths: is hepatitis C a factor? J Clin Pathol. 2002;55: 704-707.
  120. Tsui JI, Pletcher MJ, Vittinghoff E, Seal K, Gonzales R. Hepatitis C and hospital outcomes in patients admitted with alcohol-related problems. J Hepatol. 2006;44: 262-266.
  121. Novo-Veleiro I, Alvela-Suárez L, Chamorro A-J, González-Sarmiento R, Laso F-J, Marcos M. Alcoholic liver disease and hepatitis C virus infection. World J Gastroenterol. 2016;22: 1411-1420.
  122. Forrest E, Mellor J, Stanton L, Bowers M, Ryder P, Austin A, et al. Steroids or pentoxifylline for alcoholic hepatitis (STOPAH): study protocol for a randomised controlled trial. Trials. 2013;14: 262.
  123. Rambaldi A, Saconato HH, Christensen E, Thorlund K, Wetterslev J, Gluud C. Systematic review: glucocorticosteroids for alcoholic hepatitis--a Cochrane Hepato-Biliary Group systematic review with meta-analyses and trial sequential analyses of randomized clinical trials. Aliment Pharmacol Ther. 2008;27: 1167-1178.
  124. Mathurin P, Mendenhall CL, Carithers RL Jr, Ramond M-J, Maddrey WC, Garstide P, et al. Corticosteroids improve short-term survival in patients with severe alcoholic hepatitis (AH): individual data analysis of the last three randomized placebo controlled double blind trials of corticosteroids in severe AH. J Hepatol. 2002;36: 480-487.
  125. Lucey MR, Mathurin P, Morgan TR. Alcoholic hepatitis. N Engl J Med. 2009;360: 2758-2769.
  126. Louvet A, Naveau S, Abdelnour M, Ramond M-J, Diaz E, Fartoux L, et al. The Lille model: a new tool for therapeutic strategy in patients with severe alcoholic hepatitis treated with steroids. Hepatology. 2007;45: 1348-1354.
  127. Depew W, Boyer T, Omata M, Redeker A, Reynolds T. Double-blind controlled trial of prednisolone therapy in patients with severe acute alcoholic hepatitis and spontaneous encephalopathy. Gastroenterology. 1980;78: 524-529.
  128. Singal AK, Walia I, Singal A, Soloway RD. Corticosteroids and pentoxifylline for the treatment of alcoholic hepatitis: Current status. World J Hepatol. 2011;3: 205-210.
  129. Lieber CS. S-Adenosyl-L-methionine and alcoholic liver disease in animal models: implications for early intervention in human beings. Alcohol. 2002;27: 173-177.
  130. Dominguez M, Miquel R, Colmenero J, Moreno M, GarcíaPagán J-C, Bosch J, et al. Hepatic expression of CXC chemokines predicts portal hypertension and survival in patients with alcoholic hepatitis. Gastroenterol. 2009;136: 1639-1650.
  131. Gao B, Xu M. Chemokines and alcoholic hepatitis: are chemokines good therapeutic targets? Gut. 2014;63: 1683-1684.
  132. Horiguchi N, Wang L, Mukhopadhyay P, Park O, Jeong WI, Lafdil F, et al. Cell type-dependent pro- and anti-inflammatory role of signal transducer and activator of transcription 3 in alcoholic liver injury. Gastroenterology. 2008;134: 1148-1158.
  133. Støy S, Sandahl TD, Dige AK, Agnholt J, Rasmussen TK, Grønbæk H, et al. Highest frequencies of interleukin22-producing T helper cells in alcoholic hepatitis patients with a favourable short-term course. PLoS One. 2013;8: e55101.
  134. Ki SH, Park O, Zheng M, Morales-Ibanez O, Kolls JK, Bataller R, et al. Interleukin-22 treatment ameliorates alcoholic liver injury in a murine model of chronic-binge ethanol feeding: role of signal transducer and activator of transcription 3. Hepatology. 2010;52: 1291-1300.
  135. Radaeva S, Sun R, Pan H-N, Hong F, Gao B. Interleukin 22 (IL-22) plays a protective role in T cell-mediated murine hepatitis: IL-22 is a survival factor for hepatocytes via STAT3 activation. Hepatology. 2004;39: 1332-1342.
  136. Jeong W-I, Osei-Hyiaman D, Park O, Liu J, Bátkai S, Mukhopadhyay P, et al. Paracrine activation of hepatic CB1 receptors by stellate cell-derived endocannabinoids mediates alcoholic fatty liver. Cell Metab. 2008;7: 227-235.
  137. Louvet A, Teixeira-Clerc F, Chobert M-N, Deveaux V, Pavoine C, Zimmer A, et al. Cannabinoid CB2 receptors protect against alcoholic liver disease by regulating Kupffer cell polarization in mice. Hepatology. 2011;54: 1217-1226.
  138. Wang KX, Denhardt DT. Osteopontin: role in immune regulation and stress responses. Cytokine Growth Factor Rev. 2008;19: 333-345.
  139. Altamirano J, Bataller R. Alcoholic liver disease: pathogenesis and new targets for therapy. Nat Rev Gastroenterol Hepatol. 2011;8: 491-501.
  140. Sullivan S, Kirk EP, Mittendorfer B, Patterson BW, Klein S. Randomized trial of exercise effect on intrahepatic triglyceride content and lipid kinetics in nonalcoholic fatty liver disease. Hepatology. 2012;55:1738-45.
  141. Promrat K, Kleiner DE, Niemeier HM, Jackvony E, Kearns M, Wands JR, et al. Randomized controlled trial testing the effects of weight loss on nonalcoholic steatohepatitis. Hepatology. 2010;51:121-9.
  142. Harrison SA, Fecht W, Brunt EM, Neuschwander-Tetri BA. Orlistat for overweight subjects with nonalcoholic steatohepatitis: A randomized, prospective trial. Hepatology. 2009;49:80-6.
  143. Dyson J, Day C. Treatment of non-alcoholic fatty liver disease. Dig Dis. 2014;32:597-604.
  144. Zivkovic AM, German JB, Sanyal AJ. Comparative review of diets for the metabolic syndrome: implications for nonalcoholic fatty liver disease. Am J Clin Nutr. 2007;86:285-300.
  145. Musso G, Gambino R, Pacini G, De Michieli F, Cassader M. Prolonged saturated fat-induced, glucose-dependent insulinotropic polypeptide elevation is associated with adipokine imbalance and liver injury in nonalcoholic steatohepatitis: dysregulated enteroadipocyte axis as a novel feature of fatty liver. Am J Clin Nutr. 2009;89:558-67.
  146. Araya J, Rodrigo R, Videla LA, Thielemann L, Orellana M, Pettinelli P, et al. Increase in long-chain polyunsaturated fatty acid n - 6/n - 3 ratio in relation to hepatic steatosis in patients with non-alcoholic fatty liver disease. Clin Sci (Lond). 2004;106:635-43.
  147. Elizondo A, Araya J, Rodrigo R, Poniachik J, Csendes A, Maluenda F, et al. Polyunsaturated fatty acid pattern in liver and erythrocyte phospholipids from obese patients. Obesity (Silver Spring). 2007;15:24-31.
  148. Parker HM, Johnson NA, Burdon CA, Cohn JS, O'Connor HT, George J. Omega-3 supplementation and non-alcoholic fatty liver disease: a systematic review and meta-analysis. J Hepatol. 2012;56:944-51.
  149. Wong VW, Chan RS, Wong GL, Cheung BH, Chu WC, Yeung DK, et al. Community-based lifestyle modification programme for non-alcoholic fatty liver disease: a randomized controlled trial. J Hepatol. 2013;59:536-42.
  150. Kirwan JP, Solomon TP, Wojta DM, Staten MA, Holloszy JO. Effects of 7 days of exercise training on insulin sensitivity and responsiveness in type 2 diabetes mellitus. Am J Physiol Endocrinol Metab. 2009;297:E151-6.
  151. Van Der Heijden GJ, Wang ZJ, Chu Z, Toffolo G, Manesso E, Sauer PJ, et al. Strength exercise improves muscle mass and hepatic insulin sensitivity in obese youth. Med Sci Sports Exerc. 2010;42:1973-80.
  152. Thoma C, Day CP, Trenell MI. Lifestyle interventions for the treatment of non-alcoholic fatty liver disease in adults: a systematic review. J Hepatol. 2012;56:255-66.
  153. Hallsworth K, Fattakhova G, Hollingsworth KG, Thoma C, Moore S, Taylor R, et al. Resistance exercise reduces liver fat and its mediators in non-alcoholic fatty liver disease independent of weight loss. Gut. 2011;60:1278-83.
  154. Little JP, Gillen JB, Percival ME, Safdar A, Tarnopolsky MA, Punthakee Z, et al. Low-volume high-intensity interval training reduces hyperglycemia and increases muscle mitochondrial capacity in patients with type 2 diabetes. J Appl Physiol. 2011;111:1554-60.
  155. Centre for Public Health Excellence at NICE (UK); National Collaborating Centre for Primary Care (UK). Obesity: The Prevention, Identification, Assessment and Management of Overweight and Obesity in Adults and Children [Internet]. London: National Institute for Health and Clinical Excellence (UK); 2006 Dec. PMID: 22497033.
  156. Zelber-Sagi S, Nitzan-Kaluski D, Goldsmith R, Webb M, Zvibel I, Goldiner I, et al. Role of leisure-time physical activity in nonalcoholic fatty liver disease: a populationbased study. Hepatol. 2008;48:1791-8.
  157. Hsieh SD, Yoshinaga H, Muto T, Sakurai Y. Regular physical activity and coronary risk factors in Japanese men. Circulation. 1998;97:661-5.
  158. Frith J, Day CP, Robinson L, Elliott C, Jones DE, Newton JL. Potential strategies to improve uptake of exercise interventions in non-alcoholic fatty liver disease. J Hepatol. 2010;52:112-6.
  159. Centis E, Marzocchi R, Suppini A, Dalle Grave R, Villanova N, Hickman IJ, et al. The role of lifestyle change in the prevention and treatment of NAFLD. Curr Pharm Des. 2013;19:5270-9.
  160. Mummadi RR, Kasturi KS, Chennareddygari S, Sood GK. Effect of bariatric surgery on nonalcoholic fatty liver disease: systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2008;6:1396-402.
  161. Singh S, Osna NA, Kharbanda KK. Treatment options for alcoholic and non-alcoholic fatty liver disease: A review. World J Gastroenterol. 2017;23:6549-6570.
  162. Chavez-Tapia NC, Tellez-Avila FI, Barrientos-Gutierrez T, Mendez-Sanchez N, Lizardi-Cervera J, Uribe M. Bariatric surgery for non-alcoholic steatohepatitis in obese patients. Cochrane Database Syst Rev. 2010;2010:CD007340.
  163. Harrison SA, Torgerson S, Hayashi P, Ward J, Schenker S. Vitamin E and vitamin C treatment improves fibrosis in patients with nonalcoholic steatohepatitis. Am J Gastroenterol. 2003;98:2485-2490.
  164. Kugelmas M, Hill DB, Vivian B, Marsano L, McClain CJ. Cytokines and NASH: a pilot study of the effects of lifestyle modification and vitamin E. Hepatol. 2003;38:413-419.
  165. Lavine JE, Schwimmer JB, Van Natta ML, Molleston JP, Murray KF, Rosenthal P, et al. Nonalcoholic Steatohepatitis Clinical Research Network. Effect of vitamin E or metformin for treatment of nonalcoholic fatty liver disease in children and adolescents: the TONIC randomized controlled trial. JAMA. 2011;305:1659-68.
  166. Adams LA, Angulo P. Vitamins E and C for the treatment of NASH: duplication of results but lack of demonstration of efficacy. Am J Gastroenterol. 2003;98:2348-2350.
  167. Sanyal AJ, Mofrad PS, Contos MJ, Sargeant C, Luketic VA, Sterling RK, et al. A pilot study of vitamin E versus vitamin E and pioglitazone for the treatment of nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 2004;2:1107-1115.
  168. Miller ER 3rd, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ, Guallar E. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med. 2005;142:37-46.
  169. Romanelli RG, Caligiuri A, Carloni V, DeFranco R, Montalto P, Ceni E, et al. Effect of pentoxifylline on the degradation of procollagen type I produced by human hepatic stellate cells in response to transforming growth factor-beta 1. Br J Pharmacol. 1997;122:1047-54.
  170. Préaux AM, Mallat A, Rosenbaum J, Zafrani ES, Mavier P. Pentoxifylline inhibits growth and collagen synthesis of cultured human hepatic myofibroblast-like cells. Hepatology. 1997;26:315-22.
  171. Zein CO, Yerian LM, Gogate P, Lopez R, Kirwan JP, Feldstein AE, et al. Pentoxifylline improves nonalcoholic steatohepatitis: a randomized placebo-controlled trial. Hepatol. 2011;54:1610-9.
  172. Im GY, Lucey MR. Practical Concerns and Controversies in the Management of Alcoholic Hepatitis. Gastroenterol Hepatol (N Y) 2016;12:478-489.
  173. Thursz MR, Richardson P, Allison M, Austin A, Bowers M, Day CP, et al. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med. 2015;372:1619-1628.
  174. Bugianesi E, Gentilcore E, Manini R, Natale S, Vanni E, Villanova N, et al. A randomized controlled trial of metformin versus vitamin E or prescriptive diet in nonalcoholic fatty liver disease. Am J Gastroenterol. 2005;100:1082-1090.
  175. Loomba R, Lutchman G, Kleiner DE, Ricks M, Feld JJ, Borg BB, et al. Clinical trial: pilot study of metformin for the treatment of non-alcoholic steatohepatitis. Aliment Pharmacol Ther. 2009;29:172-182.
  176. Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, et al. American Gastroenterological Association; American Association for the Study of Liver Diseases; American College of Gastroenterologyh. 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 Gastroenterology. Gastroenterology. 2012 Jun;142:1592-609.
  177. Lavine JE, Schwimmer JB, Van Natta ML, Molleston JP, Murray KF, Rosenthal P, et al. Nonalcoholic Steatohepatitis Clinical Research Network. Effect of vitamin E or metformin for treatment of nonalcoholic fatty liver disease in children and adolescents: the TONIC randomized controlled trial. JAMA. 2011;305:1659-1668.
  178. Li Y, Liu L, Wang B, Wang J, Chen D. Metformin in non-alcoholic fatty liver disease: a systematic review and meta-analysis. Biomed Rep. 2013;1:57-64.
  179. Musso G, Gambino R, Cassader M, Pagano G. A meta-analysis of randomized trials for the treatment of nonalcoholic fatty liver disease. Hepatol. 2010;52:79-104.
  180. Yokohama S, Yoneda M, Haneda M, Okamoto S, Okada M, Aso K, et al. Therapeutic efficacy of an angiotensin II receptor antagonist in patients with nonalcoholic steatohepatitis. Hepatology. 2004;40:1222-1225.
  181. Basaranoglu M, Acbay O, Sonsuz A. A controlled trial of gemfibrozil in the treatment of patients with nonalcoholic steatohepatitis. J Hepatol. 1999;31:384.
  182. Laurin J, Lindor KD, Crippin JS, Gossard A, Gores GJ, Ludwig J, et al. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatol. 1996;23:1464-1467.
  183. Assy N, Grozovski M, Bersudsky I, Szvalb S, Hussein O. Effect of insulin-sensitizing agents in combination with ezetimibe, and valsartan in rats with non-alcoholic fatty liver disease. World J Gastroenterol. 2006;12:4369-4376.
  184. Zheng S, Hoos L, Cook J, Tetzloff G, Davis H Jr, van Heek M, et al. Ezetimibe improves high fat and cholesterol diet-induced non-alcoholic fatty liver disease in mice. Eur J Pharmacol. 2008;584:118-124.
  185. Akdemir A, Sahin C, Erbas O, Yeniel AO, Sendag F. Is ursodeoxycholic acid crucial for ischemia/reperfusion-induced ovarian injury in rat ovary? Arch Gynecol Obstet. 2015;292:445-50.
  186. Canbolat İP, Yiğittürk G, Erbaş O. Anti-inflammatory and anti-fibrotic effects of ursodeoxycholic acid in streptozocin-induced diabetic rats. Kocaeli Med J 2020;9;1:83-88.
  187. Erdoğan, M. A., Yiğittürk, G., Erbaş, O. The neuroprotective effects of ursodeoxycholic acid (UDCA) on experimental Parkinson model in rats. Anatomy: International Journal of Experimental & Clinical Anatomy. 2019;13.
  188. Neuman M, Angulo P, Malkiewicz I, Jorgensen R, Shear N, Dickson ER, et al. Tumor necrosis factor-alpha and transforming growth factor-beta reflect severity of liver damage in primary biliary cirrhosis. J Gastroenterol Hepatol. 2002;17:196-202.
  189. Bellentani S. Immunomodulating and anti-apoptotic action of ursodeoxycholic acid: where are we and where should we go? Eur J Gastroenterol Hepatol. 2005;17:137-140.
  190. Laurin J, Lindor KD, Crippin JS, Gossard A, Gores GJ, Ludwig J, et al. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatol. 1996;23:1464-1467.
  191. Dufour JF, Oneta CM, Gonvers JJ, Bihl F, Cerny A, Cereda JM, et al. Swiss Association for the Study of the Liver. Randomized placebo-controlled trial of ursodeoxycholic acid with vitamin e in nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 2006;4:1537-1543.
  192. Polyzos SA, Kountouras J, Mantzoros CS. Obesity and nonalcoholic fatty liver disease: from pathophysiology to therapeutics. Metabolism. 2019;92:82-97.
  193. Wang H, Wang L, Cheng Y, Xia Z, Liao Y, Cao J. Efficacy of orlistat in non-alcoholic fatty liver disease: a systematic review and meta-analysis. Biomed Rep. 2018;9:90-96.
  194. Harrison SA, Fecht W, Brunt EM, Neuschwander-Tetri BA. Orlistat for overweight subjects with nonalcoholic steatohepatitis: a randomized, prospective trial. Hepatol. 2009;49:80-86.
  195. Tuccinardi D, Farr OM, Upadhyay J, Oussaada SM, Mathew H, Paschou SA, et al. Lorcaserin treatment decreases body weight and reduces cardiometabolic risk factors in obese adults: a six-month, randomized, placebo-controlled, double-blind clinical trial. Diabetes Obes Metab. 2019;21:1487-1492.
  196. Solga SF, Diehl AM. Non-alcoholic fatty liver disease: lumen-liver interactions and possible role for probiotics. J Hepatol. 2003;38:681-687.
  197. Miele L, Valenza V, La Torre G, Montalto M, Cammarota G, Ricci R, et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatol. 2009;49:1877-1887.
  198. Aller R, De Luis DA, Izaola O, Conde R, Gonzalez Sagrado M, Primo D, et al. Effect of a probiotic on liver aminotransferases in nonalcoholic fatty liver disease patients: a double blind randomized clinical trial. Eur Rev Med Pharmacol Sci. 2011;15:1090-1095.
  199. Malaguarnera M, Vacante M, Antic T, Giordano M, Chisari G, Acquaviva R, et al. Bifidobacterium longum with fructo-oligosaccharides in patients with non alcoholic steatohepatitis. Dig Dis Sci. 2012;57:545-553.
  200. Charlton MR, Burns JM, Pedersen RA, Watt KD, Heimbach JK, Dierkhising RA. Frequency and outcomes of liver transplantation for nonalcoholic steatohepatitis in the United States. Gastroenterology. 2011;141:1249-53.
  201. Tokat Y. Hepatocellular carcinoma experience of Florence Nightingale Hospital Liver Transplantation Unit. FNG & Bilim Tranplant. Dergisi 2016; 1: 1-4.
  202. Malik SM, Devera ME, Fontes P, Shaikh O, Sasatomi E, Ahmad J. Recurrent disease following liver transplantation for nonalcoholic steatohepatitis cirrhosis. Liver Transpl. 2009;15:1843-51.