Chapter 2
Current Therapeutic Strategies for Alcoholic Liver Disease Alaa El-Din El-Sayed El-Sisi, Samia Salim Sokar, Dina Zakaria Mohamed Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
1. INTRODUCTION One of the main global public health problems is chronic alcohol consumption. Alcoholic liver disease (ALD) is caused by many critical factors. One of these factors is excessive alcohol consumption.1 A study by Schwartz and Reinus2 showed that steatosis, steatohepatitis, fibrosis, and cirrhosis are considered the main features of ALD. Alcoholic liver fibrosis can cause cirrhosis, so it is considered the cornerstone in ALD.3 The study of Wilfred de Alwis4 investigated that ethanol metabolism contributes to the pathogenesis of ALD as the formed products of its metabolism have toxic effects on the liver, and this explains the molecular mechanisms of ethanol. Almost all ingested ethanol is metabolized in the liver. There are two pathways for ethanol metabolism, namely, the oxidative and nonoxidative pathways.5,6 The oxidative pathway encompasses the alcohol dehydrogenases (ADHs) and members of the cytochrome P450 system (predominantly CYP2E1),5,7 and this pathway generates acetaldehyde. Acetaldehyde dehydrogenase (ALDH), a mitochondrial enzyme, is responsible for the metabolism of acetaldehyde to acetate, and the kinetics of this reaction is sufficiently slow to allow for the accumulation of acetaldehyde in humans or animals consuming alcohol.5,6,8 The fatty acid ethyl ester (FAEE) synthases and phospholipase D are involved in the nonoxidative pathway of ethanol metabolism and are responsible for the esterification of ethanol with fatty acids to form FAEE and phosphatidylethanol, respectively.5,6 Accumulation of NADH and the reduction of NAD+/NADH ratio are resulted from ADH and ALDH reactions and have an important effect on biochemical pathways such as glycolysis, citric acid cycle, fatty acid oxidation, and glucogenesis (Fig. 2.1). Mitochondrial electron transfer chain is responsible for reoxidizing NADH to NAD+.9,10 Different reactive oxygen species (ROS) such as superoxide anion (O2 •-), hydrogen peroxide (H2O2), and the hydroxyl radical (OH.) are formed during the electron transfer to oxygen.9 These species are unstable and rapidly react with additional electrons and protons. A small proportion can generate toxic effects as lipid peroxidation, enzymes inactivation, DNA mutations, and destruction of cell membranes.11,12 The most of these ROS are converted to water before they can damage cells.13 The microsomal ethanol oxidizing system is another metabolic system involved in ethanol metabolism and constituted by the cytochrome P450 (CYP) enzymes. These proteins are involved in the oxidation of numerous endogenous substrates such as steroids, fatty acids, and xenobiotics.14 They catalyze many different reactions to convert different chemical molecules in more polar metabolites to be excreted as monooxygenation, peroxidation, dealkylation, epoxidation, and dehalogenation. An ethanol-inducible form of P45015 catalyzes ethanol oxidation at rates much higher than other CYP enzymes. Ten percent of ethanol is oxidized to acetaldehyde by CYP2E1 physiologically,15 and induction of the microsomal system occurred during chronic alcohol abuse5,15 and an increase in CYP2E1 protein expression. During chronic ethanol intake, the increase in CYP2E1 is correlated with a decrease in proteasomal degradation, which increases CYP2E1 protein stability.16,17 The oxidation of ethanol to acetaldehyde is catalyzed by CYP2E1 and then to acetate,18 but this reaction is disadvantageous in the presence of ethanol19 as a significant amount of ROS, such as (O2 •-), H2O2, OH, and the hydroxyethyl radical (HER) are formed.20,21 Chronic ethanol consumption effects on microsomal enzymes, particularly CYP2E1, and does not influence ADH activity.6,8 So, a rise in oxygen consumption; the excessive production of free radicals; and an increase in the metabolism of ethanol, vitamin A, and testosterone resulted in ALD. Generation of ROS caused by ethanol and acetaldehyde can lead to damage to the intestinal mucosal barrier.6,22 The imbalance between free radical generation and insufficient antioxidant defense mechanisms leads to cellular oxidative stress as a decrease in glutathione (GSH), vitamin E, and phosphatidylcholine that are considered main mediators for the progression of ALD.6,8,22 Dietary Interventions in Liver Disease. https://doi.org/10.1016/B978-0-12-814466-4.00002-1 Copyright © 2019 Elsevier Inc. All rights reserved.
15
16 SECTION | I Overview of Liver Health
FIGURE 2.1 Alcohol metabolism.26
Pathophysiological changes in liver cells such as neutrophils, sinusoidal endothelial cells (SECs), Kupffer cells (KCs), hepatic stellate cells (HSCs), and hepatocytes may lead to steatosis, hepatitis, and fibrosis. The principal fibroblastic cell type within the liver is HSCs and may be activated directly by ethanol and its metabolites including acetaldehyde.23–25 Production of transforming growth factor beta-1 (TGFβ-1) and several extracellular matrix (ECM) constituents including type I collagen by HSC was induced directly by ethanol and acetaldehyde.23–25
2. PATHOGENESIS OF ALCOHOLIC LIVER DISEASE 2.1 Alcoholic Fatty Liver (Steatosis) The highly metabolic organ in the body is the liver.27 It has catabolic and anabolic capabilities and acts as a central point between the fasting and feeding states. Consuming large amounts of alcohol resulted in inadequate metabolism which leads to steatosis as endoplasmic reticulum stressed and increased fatty acid and triglyceride synthesis. Also, a large amount of fat accumulation occurs when the peroxisome proliferator–activated receptor α (PPAR-α) is downregulated or depleted as it is considered a critical point for alcohol metabolism.28,29 Alcohol consumption results in the altering of sterol regulatory element-binding proteins (SREBPs) and 50 adenosine monophosphate-activated protein kinase (AMPK) expression. Both play a significant role in regulating lipid metabolism.30,31 Downregulation of AMPK by alcohol decreases its ability to inactivate SREBP and increases adenylyl cyclase activity, contributing to steatosis.32 Dietary lipids such as saturated and unsaturated fats played an important role in liver pathology induced by alcohol.33 The early stage of ALD is the fat accumulation and it may be accompanied by inflammation, fibrosis, and cirrhosis, and hepatocellular carcinoma, and people who consumed alcohol more than of 40 g/d had steatosis.34
2.2 Alcoholic Hepatitis Alcohol intake resulted in alcoholic hepatitis (AH) which is considered a progressive inflammatory liver injury and characterized by poor liver function, ductular reaction, high levels of lipopolysaccharide (LPS), and impaired hepatocyte proliferation.35–37 These mechanisms are involved in the pathogenesis of the disease along with sterile and nonsterile inflammation.38 The essential proinflammatory cytokines involved in AH are tumor necrosis factor α (TNF-α) and interleukins (IL1 & IL6). AH has a significant short-term mortality. Stopping alcohol intake resulted in improving the prognosis in early and advanced stages.38 The main treatments for AH patients are with pentoxifylline (PTX) and prednisolone, although corticosteroids increase the risk of bleeding and infections.39
2.3 Alcoholic Fibrosis Liver fibrosis is a wound-healing response to virtually all forms of chronic liver injury, and it is characterized by excessive accumulation of collagen and other ECM proteins.40,41 The major source of the high production of ECM proteins, along with portal fibroblasts and bone marrow–derived myofibroblasts, is activated HSCs. Many cytokines, chemokines, neuroendocrine factors, angiogenic factors, and components of the innate immune system induced HSC activation and fibrogenesis after hepatocellular damage.40,41
Current Therapeutic Strategies for Alcoholic Liver Disease Chapter | 2 17
2.4 Alcoholic Cirrhosis The most frequent cause of cirrhosis in Europe is excess alcohol consumption.1 The prevalence of cirrhosis is 0.27%, corresponding to 633,323 adults in the United States in 2015.42 So, a significant cause of morbidity and mortality worldwide is liver cirrhosis. Factors that synergized the progression of the disease in addition to alcohol consumption are the lifestyle, gender, ethnic, and socioeconomic factors. Cirrhosis is more frequent, and features such as hepatocellular damage, lobular inflammation, hepatocellular ballooning degeneration, hepatocyte dropout, mega mitochondria, and the appearance of Mallory–Denk bodies are more dominant in decompensated ALD patients.43 Variceal bleeding, ascites and hepatic encephalopathy are severe complications of liver cirrhosis. As well, this cirrhosis can lead to decrease in sinusoidal space, SEC fenestrations collapse, and formation of numerous new vessels around the cirrhotic nodules bypassing the obstructed route (vascular proliferation) leading to liver architecture remodeling and portal hypertension. Finally, severe cirrhosis is considered an indicator for liver transplantation and death.44
3. CURRENT THERAPIES FOR ALCOHOLIC LIVER DISEASE 3.1 Abstinence and Lifestyle Modification Reducing alcohol consumption is a good step to improve survival rate in alcoholic cirrhotic patients, even those with decompensated liver function, and leads to the resolution of alcoholic fatty liver disease.45,46 The main problems in ALD treatment are motivating patients to follow treatment regimen, monitoring their compliance, and preventing relapse. Rehabilitation programs are important for ALD patients and maintain their calmness.47 Another published report48 showed that the active participation in Alcoholics Anonymous is the best method of encouraging ALD patients. A good step in supporting patients with alcohol addiction is recognition and treatment of comorbid psychiatric conditions.49 The method of supporting patients against alcoholic intake must be pharmacotherapy in combination with psychosocial interventions. Combination of naltrexone and acamprosate can be used for heavy drinkers.50 Disulfiram has good outcomes for alcohol-dependent individuals.51 Although disulfiram had been FDA approved, it is still widely used but less clearly confirmed by clinical trial evidence.52 Additionally, Kenna et al.48 revealed that topiramate is an effective drug in clinical trials by lowering withdrawal symptoms and enhancing the quality of life measures among alcoholic patients. Eventually, the best drug for alcoholic patients with liver cirrhosis is baclofen as reported by Addolorato et al.53 Smoking cessation and weight loss are the other considered lifestyle modifications that improve alcoholism as smoking may lead to more severe ALD and then to Hepatocellular carcinoma.54,55 Also, obesity is considered one of the important risk factors that cause fatty liver, nonalcoholic steatohepatitis, and cirrhosis and enhance the development of ALD.56
3.2 Nutritional Support and Supplements Anorexia, vomiting, maldigestion, iatrogenic causes, metabolic disturbance, hypermetabolic state, impaired protein synthesis, and malabsorption can lead to poor dietary intake and then to malnutrition that is considered a major complication of ALD.57,58 So, to regenerate hepatocyte after malnutrition, nutritional supplementations are necessary as reported by Hirsch et al.59 Symptoms of protein–energy malnutrition are muscle wasting, edema, and loss of subcutaneous fat, so a protein intake of 1.5 g/Kg and 35-40 Kcal/Kg per day is recommended for ALD patients.60 If there are folate and thiamine defi ciencies, supplementation should be considered.61,62 Zinc deficiency is an example of micronutrient deficiency in ALD, and zinc supplementation can improve ADH activity and lower CYP2E1 in animal models, enhancing ALD.63,64
3.3 Pharmacological Drugs and New Agents That Are Under Development Although patients living with ALD have mostly shown strategies to encourage abstinence from alcohol, some patients may need to be accompanied with pharmacological treatment approaches.
3.3.1 Corticosteroids Corticosteroids were considered one of the most effective drugs used for AH treatment.65 The basic mechanisms of action of corticosteroids are improving the inflammatory response by decreasing TNF-α, IL-6, and IL-866,67 and inhibiting collagen formation by suppressing the formation of acetaldehyde adduct metabolites.68 Prednisolone is one among the corticosteroid family that prescribed to treat AH with a dose of 40 mg/d for 28 d followed by tapering over 2–4 wk. Another supporting study conducted by Phillips et al.69 confirmed that corticosteroid group (prednisone 30 mg/d or methylprednisone 24 mg/d IV)
18 SECTION | I Overview of Liver Health
for 1 month showed high survival rate more than the control group (37/53) in 101 acute AH patients. Serum bilirubin is indicated as a good indicator for liver disease, so glucocorticoid can diminish it.70,71 Corticosteroids are used in severe AH, although it caused sepsis, hepatitis B, hepatorenal syndrome, and gastrointestinal (GI) bleeding as contraindications.69,72 Long-time usage of corticosteroids may increase the efficacy or show no benefits after more than 6 months.73
3.3.2 Pentoxifylline It is used as an alternative medicine for corticosteroid in AH patients when there is a contraindication to steroids, and it is used for short period of time.74,75 Although the mechanism of action of PTX is not evident, published report of Doherty et al.76 showed that 100–1000 μg/mL of PTX significantly reduced serum TNF-α levels in murine peritoneal exudate cells that were treated with endotoxin 1 μg/mL in vivo, so PTX may has anti-TNF actions. PTX and corticosteroid treatment groups or their combination showed no significant changes,77 and another metaanalysis study by Whitfield et al.78 showed that PTX decreased the hepatic mortality rate, but trial sequential analysis did not support this. Nonetheless, PTX is recommended from European and the American Association for the Study of Liver Diseases as an alternative medicine for corticosteroid in AH patients when there is a contraindication to steroids.79
3.3.3 S-Adenosyl-l-Methionine It acts as a methyl donor that is used for GSH synthesis, a major cellular antioxidant. Abnormality of hepatic gene expression in methionine and GSH metabolism that is observed in AH and cirrhotic patients resulted in decreased hepatic S-adenosyll-methionine (SAM), cysteine, and GSH levels.80 SAM supplementation can reverse liver injury and mitochondrial damage by increasing SAM concentrations as its deficiency is caused in the early stages of ALD.81 These results are in agreement with Karaa et al.82 who demonstrated that SAM can inhibit oxidative stress and TGF-α signaling pathway and then reduction in HSC activation in ethanol-lipopolysaccharide-induced liver fibrosis in rats. Large and high-quality clinical trials are needed to further prove clinical benefits of SAM in ALD as the studies of Medici et al. and Le et al.83,84 indicated that SAM does not enhance liver histopathology scores or steatosis, inflammation, and fibrosis in patients with ALD, and after examination of liver biopsies, there is no significant changes between SAM and the placebo.
3.3.4 Peroxisome Proliferator–Activated Receptor-Alpha It is a member of the nuclear receptor superfamily, mainly expressed in liver, and is responsible for genes transcription regulation that involved in oxidation, transportation of fatty acids (FAs), and exportation of free fatty acid, and alcohol intake leads to suppression of PPAR-α in hepatocytes and then inhibits FA oxidation.85 PPAR-α agonists have antiinflammatory and hypolipidemic effects by induction of FA oxidation, so these agonists are suitable for ALD patients. A new drug for ALD patients is adiponectin; it can manage the disease through PPAR-α-, TNF-α-, and sirtuin 1 (SIRT-1)-related pathways.86 Rosiglitazone is considered one of the PPAR-α agonists and can enhance hepatic adiponectin receptors (AdipoRs) in ethanol-fed mice through increasing the circulating level of adiponectin, and these mechanisms are correlated with the activation of the SIRT-1-AMPK signaling system.87
3.3.5 Carvedilol It is one among the beta-blocker family and recently used in AH patients through downregulation of fatty acid synthase and sterol regulator element binding protein 1 (SREBP-1), and upregulated PPAR-α that leads to decrease in hepatic trigylceride (TG) levels and lipid droplets within hepatocytes and also decreased HSC activation, induction of TGF-β1, and collagen and prevented ethanol-induced thickening of zone 3 vessel walls.88
3.3.6 Metformin It is an antidiabetic drug, recently used to enhance hepatic steatosis (HS) through the prevention of the upregulation of plasminogen activator inhibitor-1 and improvement of insulin resistance and liver injury by increasing PPAR-γ and adiponectin levels that lead to decrease fat accumulation and liver damage in mouse models of acute and chronic alcohol stimulation.89,90
3.3.7 Diethylcarbamazine Is the drug most widely used in the treatment of lymphatic filariasis since 1947.91 Some studies performed on vertebrates show that this drug has several direct biochemical effects on different enzyme systems, including glycolysis,
Current Therapeutic Strategies for Alcoholic Liver Disease Chapter | 2 19
folate metabolism, and activity of acetyl cholinesterase.92 Also, diethylcarbamazine (DEC) has antiinflammatory properties as a result of its interference with the arachidonic acid metabolism, which includes lipoxygenase and cyclooxygenase enzymes.93,94 A study performed in the laboratory of the authors of the present study analyzed the antiinflammatory effect of DEC on hepatic cells of alcoholic mice. The DEC-treated group, which received 50 mg/kg for 12 d (a dose equivalent to 6 mg/kg given to human preconized by OMS), significantly reduced several parameters of chronic hepatic inflammation. Histological and ultrastructural analysis of the alcoholic group showed evidence of hepatocellular damage; which was strikingly reduced in the alcoholic DEC-treated group. Also DEC treatment reduced steatosis, necrosis, and foci of inflammatory infiltrates and diminished serum AST levels and inflammatory markers such as malondialdehyde (MDA), NF-κBp65, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), vascular cell adhesion molecule-1(VCAM-1), intercellular cell adhesion molecule (ICAM-1), monocyte chemotactic peptide1(MCP-1) and its functional receptor C2 chemokine receptor (CCR2), and inducible nitric oxide synthase (iNOS) in hepatic tissue. This study found that chronic consumption of ethanol increased NF-κBp65 and targeted several proinflammatory cytokines, chemokines, and oxidases. Moreover, the administration of DEC inhibited NF-κBp65 expression, reducing hepatic injury and decreasing inflammatory markers, suggesting a potential therapeutic use in chronic inflammation.95 Another published report of Rodrigues et al.96 showed that treatment with DEC was able to reduce liver damage, collagen content, the expression of nuclear factor kappa-light-chain enhancer of activated B cells, and inflammatory markers. It also ameliorated biochemistry parameters (total cholesterol, high-density lipoprotein cholesterol, and triglyceride content and aspartate aminotransferase) and increased the expression of antiinflammatory markers (p-50 adenosine monophosphate–activated protein kinase and interleukin-10). Recently, the study of El-Sisi et al.97 evaluated that administration of DEC (50 mg/kg), concomitantly with EtOH, significantly decreased EtOHinduced increases in serum enzymatic activities of ALT, AST, and GGT levels, suggesting that DEC decreased the hepatic parenchyma injury. Also, DEC reduced hepatic MDA and NOx concentrations with a significant elevation of hepatic-reduced GSH concentration and diminished hepatic hydroxyproline, serum TGF-β1, and inflammatory markers such as IL-6 as well as significantly reduced the incidence of liver lesions induced by EtOH with a lesser score of fibrosis and necroinflammation compared with EtOH group. DEC concomitantly with EtOH showed a mild expression of α-SMA in the portal tract and moderate expression in the liver parenchyma compared with EtOH group. So, future clinical trials may demonstrate that oral administration of DEC may be suitable for the treatment of ALD and other liver diseases.
3.3.8 CYP2E1 Inhibitors CYP2E1 inhibitors prevent alcohol-induced liver damage. Polyenylphosphatidylcholine (PPC) is a mixture of polyunsaturated lecithins and is extracted from soybeans and acts as a CYP2E1 inhibitor by attenuation of hepatic oxidative stress and fibrosis.98 Another supporting study conducted by Lieber98 confirmed that large randomized controlled trials (RCTs) are needed to further prove clinical benefits of PPC in progressive liver fibrosis.99 Clomethiazole indicated to reduce CYP2E1 activity and reduced ethanol-induced hepatic injury in rats and humans.100,101
3.3.9 Prebiotics and TLR-4 Antagonists Establishing healthy gut microbiota has been put forward as a strategy in the treatment of ALD as liver and GI tract share a reciprocal relationship. Kinde et al.102 showed that LPS binds to CD14 in KC, which reacts with toll-like receptor 4 (TLR-4) to activate and release proinflammatory cytokines genes.102 Probiotics, prebiotics, and TLR-4 antagonists can adjust LPS signaling and gut microbiota to treat ALD.103 Gut-derived endotoxin/LPS may be important in the study of ALD as antibiotics or Lactobacillius can decrease the gut microflora in animal experiments.104 The modified probiotic Escherichia coli Nissle 1917 has an antiinflammatory, antioxidant, and antihyperlipidemia effects in acute alcohol exposure in rats by secretion of pyrroloquinoline quinone, which decreased lipid peroxidation and increased GSH.105
3.3.10 Antioxidants Products of lipid peroxidation were detected in both heavy drinkers and ALD patients, so oxidative stress is regarded as a cornerstone in the pathogenesis of ALD.106 After exposure of human hepatocyte (HepG2) to ethanol and acetaldehyde, metadoxine can lower lipid peroxidation and GSH depletion, so antioxidant therapy is important for ALD patients.106 Also, metadoxine prevents the increase of collagen production and TNF-α secretion in HSC.107
20 SECTION | I Overview of Liver Health
3.3.11 Others Drugs Colchicine has antifibrotic effects when patients with chronic liver cirrhosis received (1 mg/d) over a 3-year period as reported by Muntoni et al.108 Another published report of Carmichael et al.109 showed that propylthiouracil (PTU) as an antithyroid drug may also improve liver fibrosis. PTU enhances hepatic blood flow and also decreases oxidative stress in rats, so it was suggested as a curative drug to treat liver cirrhosis induced by alcohol.109
3.4 Liver Transplantation The final treatment option for ALD patients with alcoholic cirrhosis who have not responded to abstaining from alcohol is liver transplantation. For patients with decompensated liver injury and have not responded to pharmaceutical therapy, liver transplantation is the magic and saving choice.110 Relapse incidences after liver transplantation may occur in 10%–52% of patients.111,112 Finally, to avoid histological damage the transplantation should be followed after 6 months of abstinence.113,114
3.5 Natural and Herbal Medicines for the Prevention and Treatment of ALD Herbs as complementary and alternative medicine (CAM) are effective in the treatment of liver diseases, such as antagonizing fibrosis, steatosis, and hepatitis viruses, and in protecting the liver cell.115 There are several natural and herbal medicines that have been widely used and reviewed with efficacy and mechanisms. Table 2.1 summarizes the effects and mechanisms of natural and herbal medicines.
3.6 Herbal Formulas for Treatment of Alcoholic Liver Disease This review mentioned some herbal blends that have been used for ALD, and these blends have not been approved by the Food and Drug Administration as reported by Moon-Sun et al.49
3.6.1 Herbal Medicine 861 (HM 861) This medicine consists of 10 herbs including Salvia miltiorrhiza, Astragalus membranaceus, and Spatholobus suberectus.166 HM 861 is used for chronic hepatitis patients by inhibition proliferation and induction apoptosis of HSC.167 And in HSC-T6 cells, it can suppress tissue inhibitor metalloprotease 1 (TIMP-1) mRNA expression.168 Another published report of Baoen et al.169 demonstrated that this mixture showed antifibrotic activity by decreasing ALT levels to the normal range in 73% of patients with hepatitis B. These results are in agreement with Yin et al.170 and showed that HM 861 can enhance fibrosis and early cirrhosis in 52 patients with HBV and decrease hepatic inflammatory scores in fibrotic patients.
3.6.2 TJ-9 TJ-9 consists of seven herbal constituents including bupleurum root, Pinellia tuber, scutellaria root, jujube fruit, ginger rhizome, ginseng root, and Glycyrrhiza root. It is referred to as xiao-chai-hu-tang in China or Shosaiko-to in Japan and used for hepatic disorders. It shows antioxidant activity when preparation contains 7.5 g of TJ-9 with the active components baicalin and baicalein.171 The predictable mechanism of TJ-9 is inhibition of typeⅠcollagen production, α-smooth muscle actin, cell spreading, and suppression of HSC activation, but the exact mechanism is unknown.171 Additionally, Cohen et al.172 revealed that the administration of TJ-9 with interferon can be used for chronic hepatitis treatment and in cases of interstitial pneumonitis.
3.6.3 Liv-52 Liv-52 consists of seven herbs including Capparis spinosa (Himsara), Cichorium intybus (Kasani), Mandur bhasma, Solanum nigrum (Kakamachi), Terminalia arjuna (Arjuna), Cassia occidentalis (Kasamarda), Achillea millefolium (Biranjasipha), and Tamarix gallica (Jhavaka). It is an Indian ayurvedic medicine and has been used for the treatment of hepatic disorders.173 RCT from Europe showed that Liv-52 has effect on advanced cirrhosis induced by alcohol, although it was primarily used for ALD treatment.174 In contrast, Liv-52 improved PPAR-γ suppression and TNF-α expression in HepG2 cells.175
TABLE 2.1 The Effects of Natural and Herbal Medicines for the Prevention and Treatment of Alcoholic Liver Disease Herbal/Medicine Compound
Type of Model
Effective Dose
Effects
Mechanisms
References
Ethanol extract of Antrodia camphorate
Acute alcohol-fed Sprague– Dawley rats
47.8 and 95.6 mg/kg
↓(ALT; AST; ALP)
Antioxidation: ↓MDA and ↑(GSH; GPX; SOD)
116
Aqueous extract of Agrimonia eupatoria L.
Chronic alcohol-fed Sprague– Dawley rats
30 mg/kg
↓(ALT; AST)
Antioxidation: ↓MDA; ↑GSH Antiinflammation: ↓(TNF-α; IL-6; TLR4; NF-κB; MyD88; COX-2)
117
Fermented extract of barley
Chronic alcohol-fed Wistar rats
Not provided
↓(ALT; AST)
Antioxidation: ↑(GPX; SOD; CAT) and ↓MDA
118
Betaine
Acute alcohol-fed C57BL/6 mice
Not provided
↓ALT
Antioxidation: ↓(MDA; CYP2E1); ↑GSH Antiinflammation: ↓TNF-α
119
Chronic alcohol-fed Sprague– Dawley rats
200 and 400 mg/kg
↓(ALT; AST)
Antiinflammation: ↓(TNF-α; IL-18; endotoxin; TLR4)
120
Ethanol diet Wistar rats
1% (w/v) in diet
↓Hepatic ballooning and fat contents
↓(NOS; CYP2E1)
121
Anthocyanin-rich extract of Oryza sativa L. Japonica
Chronic alcohol-fed Wistar rats
500 mg/kg
↓(ALT; AST; GGT; TG; TC)
Antioxidation: ↓MDA and ↑(GSH; GST; SOD; GPX)
122
Caffeine
Chronic alcohol-fed Kunming mice
5, 10, and 25 mg/kg
↓(ALT; AST; TG; TC)
Antioxidation: ↓(ROS; MDA) and ↑(SOD; GPX) Antiinflammation: ↓(TNF-α; IL-1b; IL-6; MCP-1) FA synthesis: ↓(SREBP-1c; FAS; ACC; SCD-1)
123
Chronic alcohol-fed Sprague– Dawley rats
5, 10, and 20 mg/kg
↓(ALT; AST; HA; LN; PIIINP; CIV)
Antifibrotic: ↓(cAMP; PKA; CREB) signal pathway through adenosine A2A receptors in HSC
124
Leaf extract of Cajanus cajan Linn.
Chronic alcohol-fed Sprague– Dawley rats
50 mg/kg
↓(ALT; AST; ALP)
Antioxidation: ↑(GSH; SOD; CAT; GST)
125
Aqueous extract of Pecan nut shells (Carya illinoinensis)
Chronic alcohol-fed Wistar rats
3327 mg/kg
↓(ALT; AST; GGT)
Antioxidation: ↑(GSH; CAT; SOD)
126
Aqueous leaf extract of Cassia auriculata Linn.
Chronic alcohol-fed Wistar rats
250 and 500 mg/kg
↓(ALT; AST; ALP)
Antioxidation: ↑(SOD; CAT; GSH; Vit E; Vit C) and ↓ROS
127
Bark alcoholic extract of cinnamon
Acute alcohol-fed C57BL/6 mice
Not provided
↓TG
Antiinflammation: ↓MyD88
128
Osthole
Chronic alcohol-fed Kunming mice
40 mg/kg
↓(TG; TC)
Antioxidation: ↓MDA and ↑GSH Antiinflammation: ↓TNF-α
129
Antioxidation: ↑SOD and ↓(MDA; CYP2E1) FA β-oxidation: ↑CPT1
130
20 and 40 mg/kg
Continued
TABLE 2.1 The Effects of Natural and Herbal Medicines for the Prevention and Treatment of Alcoholic Liver Disease—cont’d Herbal/Medicine Compound
Type of Model
Effective Dose
Effects
Mechanisms
References
Curcumin
Chronic alcohol-fed Wistar rats
75 mg/kg
↓ALT
Antiinflammation: ↓(NF-κB; TNF-α; IL-12; MCP-1; COX-2)
131
Chronic alcohol-fed Sprague– Dawley rats
200 and 600 mg/kg
Antioxidation: ↓MDA Antiinflammation: ↓NF-κB
132
Alcohol (100 mM)-induced rat primary hepatocytes
0–50 μmol/L
↓(AST; MDA)
Improved GSH and heme oxygenase-1 (HO-1) induction
133
Flavonoid extract of Theobroma cacao
Chronic alcohol-fed Wistar rats
400 mg/kg
↓ALT
Antiinflammation: ↓TNF-α
134
Corn oligopeptides
Chronic alcohol-fed Wistar rats
900 mg/kg
↓(ALT; TC)
Antioxidation: ↑SOD and ↓MDA
135
Dieckol-rich phlorotannins of Ecklonia cava
Chronic alcohol-fed BALB/c mice
25 mg/kg
↓(ALT; AST; TC)
Antioxidation: ↑SOD and ↓MDA
136
Methanol extract of Gentiana manshurica Kitag.
Acute alcohol-fed C57BL/6 mice
200 mg/kg
↓(ALT; AST; TG)
Antioxidation: ↓(MDA; CYP2E1) and ↑(GSH; GPX; SOD; CAT) FA synthesis: ↓SREBP-1c
137
Ginkgo biloba extract of Ginkgo biloba
Chronic alcohol-fed Sprague– Dawley rats
200 mg/kg
↓ALT
Antioxidation: ↓MDA and ↑GSH Antiinflammation: ↓TNF-α
138
Chronic alcohol-fed Sprague– Dawley rats
96 mg/kg
↓(ALT; AST)
Antioxidation: ↑(SOD; GPX; CAT; GSH) and ↓MDA
139
Ethanol-feeding mice ethanol feeding hepatocytes (AML12 cell lines)
250 or 500 mg/kg for 4 week in mice
Improves histopathological changes and ↓TG
↓(Lipogenesis pathway; CYP2E1; 4-HNE; nitrotyrosine levels) and ↑activation of AMPK-SIRT1
140
Alcohol consumption with high fat diet mice
200 mg/kg per day for last 2 week
↓ALT and no different AST
↓(TNF-α; IL-1) and ↑IL-10
141
Catechin
Chronic alcohol-fed Wistar rats
50 mg/kg
↓ALP
Antioxidation: ↓MDA and ↑(GSH; SOD; CAT) Antiinflammation: ↓NF-κB; TNF-α
142
Green tea extract
Chronic alcohol-fed Wistar rats
300 mg/kg
↓ALT
Antiinflammation: ↓TNF-α
7
Chronic alcohol-fed Wistar rats
Not provided
↓(ALT; TG)
Antioxidation: ↓(ROS; CYP2E1) FA synthesis: ↓(SREBP-1c; FAS)
143
Male albino Wistar rats with ethanol (6 g/kg per day) for 60 d
EGCG (100 mg/kg per day) for the last of 30 day of ethanol administration
Normalization of activities of enzymatic
Antioxidants: ↓Lipid peroxidation
144
Female Sprague–Dawley rats with ethanol (56%)
EGCG (100 mg/bw)
↓(Gut leakiness; endotoxemia; lipid peroxidation)
↓(CD14; TNF-α; COX-2; INOS)
145
Male Wistar rats with ethanol for 5 week
EGCG contained diet (3 g/L) for 2 week and then ethanol EGCG diet for 5 wk
↓(AST; ALT)
FA oxidation: ↑(CTP-1; p-ACC)
146
Ginsenosides (red ginseng extract)
Epigallocatechin-3gallate
l-Theanine
Acute alcohol-fed ICR mice
100 and 200 mg/kg
↓(ALT; AST; TG)
Antioxidation: ↓MDA and ↑(GSH; SOD; CAT)
147
Semen Hoveniae extract of Hovenia dulcis Thunb.
Acute alcohol-fed Kunming mice
300 and 600 mg/kg
↓(ALT; AST; TG)
Antioxidation: ↓MDA and ↑(GSH; GST; SOD)
148
Aqueous extract of Ligularia fischeri (Ledeb.) Turcz.
Chronic alcohol-fed Sprague– Dawley rats
200 mg/kg
↓(ALT; AST; GGT)
Antioxidation: ↓MDA and ↑(GSH; SOD; GPX; CAT)
149
Bark ethanol extract of Magnolia officinalis
Chronic alcohol-fed Wistar rats
45 mg/kg
↓(ALT; TG)
Antioxidation: ↑GSH Antiinflammation: ↓TNF-α FA synthesis: ↓(SREBP1c; ACLY; FAS; SCD-1)
150
Honokiol
Chronic alcohol-fed Wistar rats
10 mg/kg
↓(ALT; TG)
Antioxidation: ↑GSH Antiinflammation: ↓TNF-α FA synthesis: ↓(SREBP-1c; ACC; FAS; ACLY; SCD-1)
151
Perillyl alcohol
Acute alcohol-fed Wistar rats
50 and 100 mg/kg
↓(ALT; AST)
Antioxidation: ↓MDA and ↑(CAT; GPX; GST) Antiinflammation: ↓(TNF-α; NF-κB)
152
Platycodi radix of Platycodon grandiflorus (Jacq.) A.DC.
Chronic alcohol-fed Sprague– Dawley rats
Not provided
↓(AST; TG)
Antioxidation: ↓CYP2E1
153
Ethanol (70%) extract of Pueraria lobata (Willd.) Ohwi
Chronic alcohol-fed Wistar rats
3 g/kg
↓(ALT; AST)
Antioxidation: ↑SOD Antiinflammation: ↓intestinal permeability
154
Tectoridin
Acute alcohol-fed C57BL/6 mice
50 and 100 mg/kg
↓(ALT; AST; TG)
Antioxidation: ↓MDA and ↑(SOD; GSH; GPX) FA β-oxidation: ↑CPT1
155
Puerarin
Acute alcohol-fed Wistar rats
200 mg/kg
Antioxidation: ↓MDA and ↑(SOD; GPX)
156
Resveratrol
Chronic alcohol-fed C57BL/6 mice
200 and 400 mg/kg
↓(TG; ALT)
Antioxidation: ↓MDA FA β-oxidation: ↑(CPT1; PGC-1a) FA synthesis: ↓(SREBP-1c; SCD-1; FAS; ACC) and ↑(AMPK; SIRT1) Antiinflammation: ↓TNF-α
157
Baicalin
Chronic alcohol-fed Sprague– Dawley rats
200 mg/kg
↓ALT
Antiinflammation: ↓(TLR4; MyD88; NF-κB; TNF-α; IL-6; COX-2)
119
Fenugreek seed polyphenol
Chronic alcohol-fed Wistar rats
200 mg/kg
Antioxidation: ↑(SOD; CAT; GPX; GSH; Vit E; Vit C)
158
Continued
TABLE 2.1 The Effects of Natural and Herbal Medicines for the Prevention and Treatment of Alcoholic Liver Disease—cont’d Herbal/Medicine Compound
Type of Model
Effective Dose
Effects
Mechanisms
References
Aqueous leaf extract of Ziziphus mauritiana
Chronic alcohol-fed Wistar rats
200 and 400 mg/kg
↓(ALT; AST)
Antioxidation: ↑GSH
159
Silymarin (Silybum marianum)
RCT, double-blind, 170 patients with cirrhosis
140 mg/d for three times orally
↓Rate of mortality
Alcohol-induced baboons (50% of calories)
0.84 mg/calorie for 36 mo
Improve histologic stage of fibrosis
↓Collagen I and (I) procollagen
160
Alcohol- and high fat–induced rats
100, 150, 200 mg/kg/d for 6 week
↓(ALT; AST; hepatic fat contents)
↓(NF-κB p65; ICAM-1; IL-6) were found in silymarin groups (150 mg/kg, 200 mg)
161
Vitamin E and C
Malnourished rats with ethanol
Vitamin E and C: vitamin E (15 mg/kg); vitamin C (10 mg/kg); single and combined treatments
↓(GPx; hepatic fibrosis; hepatomegaly hepatic necroinflammation)
Glycyrrhizin (Glycyrrhiza glabra)
Ethanol-CCl4-induced male SD rats
IP injections of potenlin (acquired from Hai Ning Pharmaceutical Co., Zhe Jiang, China)
↓ALT
Normalized NF-κB binding activity
163
Hesperidin
Chronic alcohol-fed Wistar rats
200 mg/kg
↓(ALT; AST; GGT)
Antioxidation: ↓(NO; MDA) and ↑GSH Antiinflammation: ↓IL-6 Antifibrotic effect: ↓(TGF-b1; α-SMA; hepatic histopathology scoring and 4-hydroxyproline content)
97
Lemon juice
Chronic alcohol-fed C57BL/6 mice
Different concentrations (high dose 1:1 [m/v], medium dose 1:5, and low dose 1:10)
↓(ALT; AST; TG)
Antioxidation: ↓(MDA) ↓Hepatic histopathology
164
Jujube honey
Chronic alcohol-fed Kunming mice
Different doses (high dose 27.0 g per kg twice daily and low dose 13.5 g per kg twice daily)
↓(ALT; AST)
Antioxidation: ↓(8-OHdG; MDA) and ↑(GSH-Px) ↓Hepatic histopathology
165
65
162
4-HNE, 4-Hydroxynonenal; 8-OHdG, 8-hydroxy-2’–deoxyguanosine; ACC, Acetyl-CoA carboxylase; ACYL, Acyl-CoA; ALP, Alkaline phosphatase; ALT, Alanine Aminotransferase; AMPK, AMP-activated protein kinase; AST, Aspartate Aminotransferase; cAMP, Cyclic adenosine monophosphate; CAT, Choline acetyltransferase; CD, cluster of differentiation; CIV, collagen type IV; COX, cyclooxygenase; CPT, Carnitine palmitoyltransferase; CREB, cAMP response element-binding protein; CYP, Cytochrome P450; EGCG, Epigallocatechin gallate; FA, Fatty acid; FAS, Fas ligand; GGT, Gamma-Glutamyl Transferase; GPX, Glutathione peroxidase; GSH, Glutathione; GST, Glutathione S-transferase; HA, hyaluronic acid; HSC, Hepatic stellate cells; ICAM-1, Intercellular Adhesion Molecule 1; IL, Interleukin; INOS, Inducible nitric oxide synthase; LN, laminin; MCP, Monocyte Chemoattractant Protein; MDA, Malondialdehyde; MyD, Myeloid differentiation primary response; NF-κB, Nuclear factor kappa B; NO, Nitric oxide,TGF-b1, Transforming growth factor beta 1; NOS, Nitric oxide synthase; PGC-1a, Peroxisome proliferator-activated receptor gamma coactivator 1-alpha; PIIINP, N-Terminal Propeptide of Type III Collagen; PKA, Protein kinase A; ROS, Reactive oxygen species; SCD-1, Stearoyl-CoA Desaturase-1; SOD, Superoxide dismutase; SREBP, Sterol regulatory element-binding protein; SREBP-1c, Sterol regulatory element-binding protein 1; TC, Total cholesterol; TG, triglyceride; TLR, Toll like receptor; TNF, Tumor necrosis factor; α-SMA, Alpha smooth muscle actin.
Current Therapeutic Strategies for Alcoholic Liver Disease Chapter | 2 25
3.7 The Combination Therapies of Drugs and Natural Agents 3.7.1 Magnesium Isoglycyrrhizinate Injection It was used for the treatment of chronic liver diseases and contains glycyrrhizin176 as well as inhibits the progress of pulmonary fibrosis.177 The double action of glycyrrhizin and matrine leads to a decrease in collagen I and HA levels secreted by HSC as compared with each drug alone. Also, this combination improves hepatic histological analysis in vitro and in vivo more effectively in CCL4-induced liver fibrosis as reported by Zhao et al.156
3.7.2 Silymarin and S-Adenosyl-l-Methionine Silymarin and SAM combination was evaluated in ALD markets with much promise.178
3.7.3 Diethylcarbamazine and Hesperidin A study performed in the laboratory by El-Sisi et al.97 analyzed the antifibrotic effects of diethylcarbamazine combined with hesperidin against ethanol-induced liver fibrosis in rats. The results of this study clearly indicate that DEC when administrated in combination with HDN has a protective effect against EtOH-induced liver fibrosis, mainly via inhibition of hepatic oxidative stress and augmentation of antioxidant defenses, attenuating the activation of HSCs via reduction of αSMA expression in liver, inhibiting the fibrogenesis and proliferation of activated HSCs response via inhibiting the release of TGF-β1, and inhibiting proinflammatory cytokines as IL-6. Overall, the combination of DEC with HDN produced augmented antifibrotic, antiinflammatory, and antioxidant effects compared with individual drugs. There have been some cases of side effects and hepatotoxicity caused by herbal medicines.179 Interstitial pneumonia occurred in chronic hepatitis patients after administration of xiao-chai-hu-tang, alone or in combination with interferon.180 Although there are beneficial effects of CAMs and conventional drug combinations, there are some herb–drug interactions that may result in toxicity in certain cases such as silymarin plus indinavir combination in AIDS patients as reported by DiCenzo et al.181
ENDNOTES 1. As an initial therapy of ALD, abstinence from alcohol accompanied with basic lifestyle modifications is appropriate. 2. When the injury becomes decompensated, pharmacological therapy should be accompanied to reverse the more severe stages of ALD. 3. The available synthetic drugs to treat ALD cause further damage to the liver, so increasing attention has been paid to herbal medicines as a newly emerging treatment strategy for ALD. 4. Combination of pharmaceutical drugs with naturally occurring agents may offer an optimal management for liver disease.
REFERENCES 1. Rehm J, Samokhvalov AV, Shield KD. Global burden of alcoholic liver diseases. J Hepatol 2013;59(1):160–8. 2. Schwartz JM, Reinus JF. Prevalence and natural history of alcoholic liver disease. Clin Liver Dis 2012;16(4):659–66. 3. Duddempudi AT. Immunology in alcoholic liver disease. Clin Liver Dis 2012;16(4):687–98. 4. Wilfred de Alwis NM, Day CP. Genetics of alcoholic liver disease and nonalcoholic fatty liver disease. Semin Liver Dis 2007;27(1):44–54. 5. Lieber CS. Cytochrome P-4502E1: its physiological and pathological role. Physiol Rev 1997;77(2):517–44. 6. Lieber CS. Metabolism of alcohol. Clin Liver Dis 2005;9(1):1–35. 7. Arteel GE, Uesugi T, Bevan LN, Gabele E, Wheeler MD, McKim SE, et al. Green tea extract protects against early alcohol-induced liver injury in rats. Biol Chem 2002;383(3–4):663–70. 8. Lieber CS. Alcoholic fatty liver: its pathogenesis and mechanism of progression to inflammation and fibrosis. Alcohol 2004;34(1):9–19. 9. Bondy SC. Ethanol toxicity and oxidative stress. Toxicol Lett 1992;63(3):231–41. 10. Bailey SM, Cunningham CC. Contribution of mitochondria to oxidative stress associated with alcoholic liver disease. Free Radic Biol Med 2002;32(1):11–6. 11. de Groot H. Reactive oxygen species in tissue injury. Hepatogastroenterology 1994;41(4):328–32. 12. Toyokuni S. Reactive oxygen species-induced molecular damage and its application in pathology. Pathol Int 1999;49(2):91–102. 13. Chance B, Sies H, Boveris A. Hydroperoxide metabolism in mammalian organs. Physiol Rev 1979;59(3):527–605. 14. Guengerich FP, Beaune PH, Umbenhauer DR, Churchill PF, Bork RW, Dannan GA, et al. Cytochrome P-450 enzymes involved in genetic polymorphism of drug oxidation in humans. Biochem Soc Trans 1987;15(4):576–8. 15. Lieber CS, DeCarli LM. The role of the hepatic microsomal ethanol oxidizing system (MEOS) for ethanol metabolism in vivo. J Pharmacol Exp Ther 1972;181(2):279–87.
26 SECTION | I Overview of Liver Health
16. Donohue Jr TM, Cederbaum AI, French SW, Barve S, Gao B, Osna NA. Role of the proteasome in ethanol-induced liver pathology. Alcohol Clin Exp Res 2007;31(9):1446–59. 17. Osna NA, Donohue Jr TM. Implication of altered proteasome function in alcoholic liver injury. World J Gastroenterol 2007;13(37):4931–7. 18. Terelius Y, Norsten-Hoog C, Cronholm T, Ingelman-Sundberg M. Acetaldehyde as a substrate for ethanol-inducible cytochrome P450 (CYP2E1). Biochem Biophys Res Commun 1991;179(1):689–94. 19. Wu YS, Salmela KS, Lieber CS. Microsomal acetaldehyde oxidation is negligible in the presence of ethanol. Alcohol Clin Exp Res 1998;22(5):1165–9. 20. Brooks PJ. DNA damage, DNA repair, and alcohol toxicity–a review. Alcohol Clin Exp Res 1997;21(6):1073–82. 21. Lu Y, Cederbaum AI. CYP2E1 and oxidative liver injury by alcohol. Free Radic Biol Med 2008;44(5):723–38. 22. Nieto N, Friedman SL, Cederbaum AI. Stimulation and proliferation of primary rat hepatic stellate cells by cytochrome P450 2E1-derived reactive oxygen species. Hepatology 2002;35(1):62–73. 23. Greenwel P. Acetaldehyde-mediated collagen regulation in hepatic stellate cells. Alcohol Clin Exp Res 1999;23(5):930–3. 24. Friedman SL. Stellate cell activation in alcoholic fibrosis–an overview. Alcohol Clin Exp Res 1999;23(5):904–10. 25. Svegliati-Baroni G, Inagaki Y, Rincon-Sanchez AR, Else C, Saccomanno S, Benedetti A, et al. Early response of alpha2(I) collagen to acetaldehyde in human hepatic stellate cells is TGF-beta independent. Hepatology 2005;42(2):343–52. 26. Ceni E, Crabb DW, Foschi M, Mello T, Tarocchi M, Patussi V, et al. Acetaldehyde inhibits PPARgamma via H2O2-mediated c-Abl activation in human hepatic stellate cells. Gastroenterology 2006;131(4):1235–52. 27. Viollet B, Guigas B, Leclerc J, Hebrard S, Lantier L, Mounier R, et al. AMP-activated protein kinase in the regulation of hepatic energy metabolism: from physiology to therapeutic perspectives. Acta Physiol (Oxf) 2009;196(1):81–98. 28. Zeng T, Zhang CL, Song FY, Zhao XL, Xie KQ. Garlic oil alleviated ethanol-induced fat accumulation via modulation of SREBP-1, PPAR-alpha, and CYP2E1. Food Chem Toxicol 2012;50(3–4):485–91. 29. Bala S, Csak T, Saha B, Zatsiorsky J, Kodys K, Catalano D, et al. The pro-inflammatory effects of miR-155 promote liver fibrosis and alcoholinduced steatohepatitis. J Hepatol 2016;64(6):1378–87. 30. Altamirano J, Bataller R. Alcoholic liver disease: pathogenesis and new targets for therapy. Nat Rev Gastroenterol Hepatol 2011;8(9):491–501. 31. Zhang M, Wang C, Wang C, Zhao H, Zhao C, Chen Y, et al. Enhanced AMPK phosphorylation contributes to the beneficial effects of Lactobacillus rhamnosus GG supernatant on chronic-alcohol-induced fatty liver disease. J Nutr Biochem 2015;26(4):337–44. 32. Garcia-Villafranca J, Guillen A, Castro J. Ethanol consumption impairs regulation of fatty acid metabolism by decreasing the activity of AMPactivated protein kinase in rat liver. Biochimie 2008;90(3):460–6. 33. Kirpich IA, Miller ME, Cave MC, Joshi-Barve S, McClain CJ. Alcoholic liver disease: update on the role of dietary fat. Biomolecules 2016;6(1):1. 34. Stickel F, Moreno C, Hampe J, Morgan MY. The genetics of alcohol dependence and alcohol-related liver disease. J Hepatol 2017;66(1):195–211. 35. Sancho-Bru P, Altamirano J, Rodrigo-Torres D, Coll M, Millan C, Jose Lozano J, et al. Liver progenitor cell markers correlate with liver damage and predict short-term mortality in patients with alcoholic hepatitis. Hepatology 2012;55(6):1931–41. 36. Dubuquoy L, Louvet A, Lassailly G, Truant S, Boleslawski E, Artru F, et al. Progenitor cell expansion and impaired hepatocyte regeneration in explanted livers from alcoholic hepatitis. Gut 2015;64(12):1949–60. 37. Odena G, Chen J, Lozano JJ, Altamirano J, Rodrigo-Torres D, Affo S, et al. LPS-TLR4 pathway mediates ductular cell expansion in alcoholic hepatitis. Sci Rep 2016;6:35610. 38. Laursen TL, Stoy S, Deleuran B, Vilstrup H, Gronbaek H, Sandahl TD. The damage-associated molecular pattern HMGB1 is elevated in human alcoholic hepatitis, but does not seem to be a primary driver of inflammation. APMIS 2016;124(9):741–7. 39. Garcia-Saenz-de-Sicilia M, Duvoor C, Altamirano J, Chavez-Araujo R, Prado V, de Lourdes Candolo-Martinelli A, et al. A Day-4 Lille model predicts response to corticosteroids and mortality in severe alcoholic hepatitis. Am J Gastroenterol 2016;112(2):306–15. 40. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest 2005;115(2):209–18. 41. Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology 2008;134(6):1655–69. 42. Scaglione S, Kliethermes S, Cao G, Shoham D, Durazo R, Luke A, et al. The epidemiology of cirrhosis in the United States: a population-based study. J Clin Gastroenterol 2015;49(8):690–6. 43. Lackner C, Spindelboeck W, Haybaeck J, Douschan P, Rainer F, Terracciano L, et al. Histological parameters and alcohol abstinence determine long-term prognosis in patients with alcoholic liver disease. J Hepatol 2017;66(3):610–8. 44. Lemoinne S, Cadoret A, Rautou PE, El Mourabit H, Ratziu V, Corpechot C, et al. Portal myofibroblasts promote vascular remodeling underlying cirrhosis formation through the release of microparticles. Hepatology 2015;61(3):1041–55. 45. Heron M, Hoyert DL, Murphy SL, Xu J, Kochanek KD, Tejada-Vera B. Deaths: final data for 2006. Natl Vital Stat Rep 2009;57(14):1–134. 46. Sofair AN, Barry V, Manos MM, Thomas A, Zaman A, Terrault NA, et al. The epidemiology and clinical characteristics of patients with newly diagnosed alcohol-related liver disease: results from population-based surveillance. J Clin Gastroenterol 2010;44(4):301–7. 47. Miller WR, Walters ST, Bennett ME. How effective is alcoholism treatment in the United States? J Stud Alcohol 2001;62(2):211–20. 48. Kaner EF, Dickinson HO, Beyer F, Pienaar E, Schlesinger C, Campbell F, et al. The effectiveness of brief alcohol interventions in primary care settings: a systematic review. Drug Alcohol Rev 2009;28(3):301–23. 49. Moos RH, King MJ, Patterson MA. Outcomes of residential treatment of substance abuse in hospital- and community-based programs. Psychiatr Serv 1996;47(1):68–74. 50. Bouza C, Angeles M, Munoz A, Amate JM. Efficacy and safety of naltrexone and acamprosate in the treatment of alcohol dependence: a systematic review. Addiction 2004;99(7):811–28. 51. Garbutt JC, West SL, Carey TS, Lohr KN, Crews FT. Pharmacological treatment of alcohol dependence: a review of the evidence. JAMA 1999;281(14):1318–25. 52. Williams SH. Medications for treating alcohol dependence. Am Fam Physician 2005;72(9):1775–80.
Current Therapeutic Strategies for Alcoholic Liver Disease Chapter | 2 27
53. 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(9603):1915–22. 54. Klatsky AL, Armstrong MA. Alcohol, smoking, coffee, and cirrhosis. Am J Epidemiol 1992;136(10):1248–57. 55. Corrao G, Lepore AR, Torchio P, Valenti M, Galatola G, D’Amicis A, et al. The effect of drinking coffee and smoking cigarettes on the risk of cirrhosis associated with alcohol consumption. A case-control study. Provincial Group for the Study of Chronic Liver Disease. Eur J Epidemiol 1994;10(6):657–64. 56. Naveau S, Giraud V, Borotto E, Aubert A, Capron F, Chaput JC. Excess weight risk factor for alcoholic liver disease. Hepatology 1997;25(1):108–11. 57. McClain CJ, Barve SS, Barve A, Marsano L. Alcoholic liver disease and malnutrition. Alcohol Clin Exp Res 2011;35(5):815–20. 58. Singal AK, Charlton MR. Nutrition in alcoholic liver disease. Clin Liver Dis 2012;16(4):805–26. 59. Hirsch S, de la Maza MP, Gattas V, Barrera G, Petermann M, Gotteland M, et al. Nutritional support in alcoholic cirrhotic patients improves host defenses. J Am Coll Nutr 1999;18(5):434–41. 60. Frazier TH, Stocker AM, Kershner NA, Marsano LS, McClain CJ. Treatment of alcoholic liver disease. Therap Adv Gastroenterol 2011;4(1):63–81. 61. Kang YJ, Zhou Z. Zinc prevention and treatment of alcoholic liver disease. Mol Aspects Med 2005;26(4–5):391–404. 62. Hanje AJ, Fortune B, Song M, Hill D, McClain C. The use of selected nutrition supplements and complementary and alternative medicine in liver disease. Nutr Clin Pract 2006;21(3):255–72. 63. Das I, Burch RE, Hahn HK. Effects of zinc deficiency on ethanol metabolism and alcohol and aldehyde dehydrogenase activities. J Lab Clin Med 1984;104(4):610–7. 64. Zhou Z, Wang L, Song Z, Saari JT, McClain CJ, Kang YJ. Zinc supplementation prevents alcoholic liver injury in mice through attenuation of oxidative stress. Am J Pathol 2005;166(6):1681–90. 65. Ferenci P, Dragosics B, Dittrich H, Frank H, Benda L, Lochs H, et al. Randomized controlled trial of silymarin treatment in patients with cirrhosis of the liver. J Hepatol 1989;9(1):105–13. 66. Taieb J, Mathurin P, Elbim C, Cluzel P, Arce-Vicioso M, Bernard B, et al. Blood neutrophil functions and cytokine release in severe alcoholic hepatitis: effect of corticosteroids. J Hepatol 2000;32(4):579–86. 67. Spahr L, Rubbia-Brandt L, Pugin J, Giostra E, Frossard JL, Borisch B, et al. Rapid changes in alcoholic hepatitis histology under steroids: correlation with soluble intercellular adhesion molecule-1 in hepatic venous blood. J Hepatol 2001;35(5):582–9. 68. Tome S, Lucey MR. Review article: current management of alcoholic liver disease. Aliment Pharmacol Ther 2004;19(7):707–14. 69. Phillips M, Curtis H, Portmann B, Donaldson N, Bomford A, O’Grady J. Antioxidants versus corticosteroids in the treatment of severe alcoholic hepatitis–a randomised clinical trial. J Hepatol 2006;44(4):784–90. 70. Mathurin P, Abdelnour M, Ramond MJ, Carbonell N, Fartoux L, Serfaty L, et al. Early change in bilirubin levels is an important prognostic factor in severe alcoholic hepatitis treated with prednisolone. Hepatology 2003;38(6):1363–9. 71. Mathurin P, Beuzin F, Louvet A, Carrie-Ganne N, Balian A, Trinchet JC, et al. Fibrosis progression occurs in a subgroup of heavy drinkers with typical histological features. Aliment Pharmacol Ther 2007;25(9):1047–54. 72. Lucey MR, Mathurin P, Morgan TR. Alcoholic hepatitis. N Engl J Med 2009;360(26):2758–69. 73. Ramond MJ, Poynard T, Rueff B, Mathurin P, Theodore C, Chaput JC, et al. A randomized trial of prednisolone in patients with severe alcoholic hepatitis. N Engl J Med 1992;326(8):507–12. 74. Akriviadis E, Botla R, Briggs W, Han S, Reynolds T, Shakil O. Pentoxifylline improves short-term survival in severe acute alcoholic hepatitis: a double-blind, placebo-controlled trial. Gastroenterology 2000;119(6):1637–48. 75. De B, Mandal S, Sau D, Mani S, Chatterjee S, Mondal S, et al. Pentoxifylline plus prednisolone versus pentoxifylline only for severe alcoholic hepatitis: a randomized controlled clinical trial. Ann Med Health Sci Res 2014;4(5):810–6. 76. Doherty GM, Jensen JC, Alexander HR, Buresh CM, Norton JA. Pentoxifylline suppression of tumor necrosis factor gene transcription. Surgery 1991;110(2):192–8. 77. Parker R, Armstrong MJ, Corbett C, Rowe IA, Houlihan DD. Systematic review: pentoxifylline for the treatment of severe alcoholic hepatitis. Aliment Pharmacol Ther 2013;37(9):845–54. 78. Whitfield K, Rambaldi A, Wetterslev J, Gluud C. Pentoxifylline for alcoholic hepatitis. Cochrane Database Syst Rev 2009;(4):Cd007339. 79. O’Shea RS, Dasarathy S, McCullough AJ. Alcoholic liver disease. Am J Gastroenterol 2010;105(1). 14–32; quiz 3. 80. Lee TD, Sadda MR, Mendler MH, Bottiglieri T, Kanel G, Mato JM, et al. Abnormal hepatic methionine and glutathione metabolism in patients with alcoholic hepatitis. Alcohol Clin Exp Res 2004;28(1):173–81. 81. Lieber CS. S-Adenosyl-l-methionine and alcoholic liver disease in animal models: implications for early intervention in human beings. Alcohol 2002;27(3):173–7. 82. Karaa A, Thompson KJ, McKillop IH, Clemens MG, Schrum LW. S-adenosyl-l-methionine attenuates oxidative stress and hepatic stellate cell activation in an ethanol-LPS-induced fibrotic rat model. Shock 2008;30(2):197–205. 83. Medici V, Virata MC, Peerson JM, Stabler SP, French SW, Gregory 3rd JF, et al. S-adenosyl-l-methionine treatment for alcoholic liver disease: a double-blinded, randomized, placebo-controlled trial. Alcohol Clin Exp Res 2011;35(11):1960–5. 84. Le MD, Enbom E, Traum PK, Medici V, Halsted CH, French SW. Alcoholic liver disease patients treated with S-adenosyl-l-methionine: an in-depth look at liver morphologic data comparing pre and post treatment liver biopsies. Exp Mol Pathol 2013;95(2):187–91. 85. Wagner M, Zollner G, Trauner M. Nuclear receptors in liver disease. Hepatology 2011;53(3):1023–34. 86. Rogers CQ, Ajmo JM, You M. Adiponectin and alcoholic fatty liver disease. IUBMB Life 2008;60(12):790–7. 87. Shen Z, Liang X, Rogers CQ, Rideout D, You M. Involvement of adiponectin-SIRT1-AMPK signaling in the protective action of rosiglitazone against alcoholic fatty liver in mice. Am J Physiol Gastrointest Liver Physiol 2010;298(3):G364–74.
28 SECTION | I Overview of Liver Health
88. Liu J, Takase I, Hakucho A, Okamura N, Fujimiya T. Carvedilol attenuates the progression of alcohol fatty liver disease in rats. Alcohol Clin Exp Res 2012;36(9):1587–99. 89. Bergheim I, Guo L, Davis MA, Lambert JC, Beier JI, Duveau I, et al. Metformin prevents alcohol-induced liver injury in the mouse: critical role of plasminogen activator inhibitor-1. Gastroenterology 2006;130(7):2099–112. 90. Zhu Z, Jiang Z, Zhou J, Zhou D, Wang W, Zhao C, et al. Involvement of insulin resistance in the protective effect of metformin against alcoholic liver injury. Alcohol Clin Exp Res 2014;38(6):1510–9. 91. Hewitt RI, White E, et al. Experimental chemotherapy of filariasis; effect of piperazine derivatives against naturally acquired filarial infections in cotton rats and dogs. J Lab Clin Med 1947;32(11):1304–13. 92. Subrahmanyam D. Antifilarials and their mode of action. Ciba Found Symp 1987;127:246–64. 93. Maizels RM, Denham DA. Diethylcarbamazine (DEC): immunopharmacological interactions of an anti-filarial drug. Parasitology 1992;(105 Suppl.):S49–60. 94. McGarry HF, Plant LD, Taylor MJ. Diethylcarbamazine activity against Brugia malayi microfilariae is dependent on inducible nitric-oxide synthase and the cyclooxygenase pathway. Filaria J 2005;4:4. 95. Santos Rocha SW, Silva BS, Gomes FO, Soares e Silva AK, Raposo C, Barbosa KP, et al. Effect of diethylcarbamazine on chronic hepatic inflammation induced by alcohol in C57BL/6 mice. Eur J Pharmacol 2012;689(1–3):194–203. 96. Rodrigues GB, Rocha SW, Dos Santos LA, de Oliveira WH, Gomes FO, de Franca ME, et al. Diethylcarbamazine: possible therapeutic alternative in the treatment of alcoholic liver disease in C57BL/6 mice. Clin Exp Pharmacol Physiol 2015;42(4):369–79. 97. El-Sisi AEE, Sokar SS, Shebl AM, Mohamed DZ. Antifibrotic effect of diethylcarbamazine combined with hesperidin against ethanol induced liver fibrosis in rats. Biomed Pharmacother 2017;89:1196–206. 98. Lieber CS. Microsomal ethanol-oxidizing system (MEOS): the first 30 years (1968–1998)–a review. Alcohol Clin Exp Res 1999;23(6):991–1007. 99. Lieber CS, Weiss DG, Groszmann R, Paronetto F, Schenker II S. Veterans Affairs Cooperative Study of polyenylphosphatidylcholine in alcoholic liver disease. Alcohol Clin Exp Res 2003;27(11):1765–72. 100. Gebhardt AC, Lucas D, Menez JF, Seitz HK. Chlormethiazole inhibition of cytochrome P450 2E1 as assessed by chlorzoxazone hydroxylation in humans. Hepatology 1997;26(4):957–61. 101. Gouillon Z, Lucas D, Li J, Hagbjork AL, French BA, Fu P, et al. Inhibition of ethanol-induced liver disease in the intragastric feeding rat model by chlormethiazole. Proc Soc Exp Biol Med 2000;224(4):302–8. 102. Kinde H, Bettey RL, Ardans A, Galey FD, Daft BM, Walker RL, et al. Clostridium botulinum type-C intoxication associated with consumption of processed alfalfa hay cubes in horses. J Am Vet Med Assoc 1991;199(6):742–6. 103. Mencin A, Kluwe J, Schwabe RF. Toll-like receptors as targets in chronic liver diseases. Gut 2009;58(5):704–20. 104. Nanji AA, Khettry U, Sadrzadeh SM. Lactobacillus feeding reduces endotoxemia and severity of experimental alcoholic liver (disease). Proc Soc Exp Biol Med 1994;205(3):243–7. 105. Singh AK, Pandey SK, Naresh Kumar G. Pyrroloquinoline quinone-secreting probiotic Escherichia coli Nissle 1917 ameliorates ethanol-induced oxidative damage and hyperlipidemia in rats. Alcohol Clin Exp Res 2014;38(7):2127–37. 106. Clot P, Tabone M, Arico S, Albano E. Monitoring oxidative damage in patients with liver cirrhosis and different daily alcohol intake. Gut 1994;35(11):1637–43. 107. Gutierrez-Ruiz MC, Bucio L, Correa A, Souza V, Hernandez E, Gomez-Quiroz LE, et al. Metadoxine prevents damage produced by ethanol and acetaldehyde in hepatocyte and hepatic stellate cells in culture. Pharmacol Res 2001;44(5):431–6. 108. Muntoni S, Rojkind M, Muntoni S. Colchicine reduces procollagen III and increases pseudocholinesterase in chronic liver disease. World J Gastroenterol 2010;16(23):2889–94. 109. Carmichael FJ, Orrego H, Saldivia V, Israel Y. Effect of propylthiouracil on the ethanol-induced increase in liver oxygen consumption in awake rats. Hepatology 1993;18(2):415–21. 110. Dureja P, Lucey MR. The place of liver transplantation in the treatment of severe alcoholic hepatitis. J Hepatol 2010;52(5):759–64. 111. Tang H, Boulton R, Gunson B, Hubscher S, Neuberger J. Patterns of alcohol consumption after liver transplantation. Gut 1998;43(1):140–5. 112. Pageaux GP, 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(5):629–34. 113. 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 Transplant Surg 1997;3(6):628–37. 114. Tome S, Martinez-Rey C, Gonzalez-Quintela A, Gude F, Brage A, Otero E, et al. Influence of superimposed alcoholic hepatitis on the outcome of liver transplantation for end-stage alcoholic liver disease. J Hepatol 2002;36(6):793–8. 115. Wang XB, Feng Y, Wang N, Cheung F, Wong CW. Recent progress on anti-liver fibrosis candidates in patents of herbal medicinal products. Recent Pat Food Nutr Agric 2012;4(2):91–106. 116. Lu ZM, Tao WY, Xu HY, Ao ZH, Zhang XM, Xu ZH. Further studies on the hepatoprotective effect of Antrodia camphorata in submerged culture on ethanol-induced acute liver injury in rats. Nat Prod Res 2011;25(7):684–95. 117. Yoon SJ, Koh EJ, Kim CS, Zee OP, Kwak JH, Jeong WJ, et al. Agrimonia eupatoria protects against chronic ethanol-induced liver injury in rats. Food Chem Toxicol 2012;50(7):2335–41. 118. Giriwono P, Hashimoto T, Ohsaki Y, Shirakawa H, Hokazono H, Komai M. Extract of fermented barley attenuates chronic alcohol induced liver damage by increasing antioxidative activities. Food Res Int 2010;42(1):118–24. 119. Kim SJ, Jung YS, Kwon DY, Kim YC. Alleviation of acute ethanol-induced liver injury and impaired metabolomics of S-containing substances by betaine supplementation. Biochem Biophys Res Commun 2008;368(4):893–8.
Current Therapeutic Strategies for Alcoholic Liver Disease Chapter | 2 29
120. Shi QZ, Wang LW, Zhang W, Gong ZJ. Betaine inhibits toll-like receptor 4 expression in rats with ethanol-induced liver injury. World J Gastroenterol 2010;16(7):897–903. 121. Kharbanda KK, Todero SL, King AL, Osna NA, McVicker BL, Tuma DJ, et al. Betaine treatment attenuates chronic ethanol-induced hepatic steatosis and alterations to the mitochondrial respiratory chain proteome. Int J Hepatol 2012;2012:962183. 122. Hou Z, Qin P, Ren G. Effect of anthocyanin-rich extract from black rice (Oryza sativa L. Japonica) on chronically alcohol-induced liver damage in rats. J Agric Food Chem 2010;58(5):3191–6. 123. Lv X, Chen Z, Li J, Zhang L, Liu H, Huang C, et al. Caffeine protects against alcoholic liver injury by attenuating inflammatory response and oxidative stress. Inflamm Res 2010;59(8):635–45. 124. Wang Q, Dai X, Yang W, Wang H, Zhao H, Yang F, et al. Caffeine protects against alcohol-induced liver fibrosis by dampening the cAMP/PKA/ CREB pathway in rat hepatic stellate cells. Int Immunopharmacol 2015;25(2):340–52. 125. Kundu R, Dasgupta S, Biswas A, Bhattacharya A, Pal BC, Bandyopadhyay D, et al. Cajanus cajan Linn. (Leguminosae) prevents alcohol-induced rat liver damage and augments cytoprotective function. J Ethnopharmacol 2008;118(3):440–7. 126. Muller LG, Pase CS, Reckziegel P, Barcelos RC, Boufleur N, Prado AC, et al. Hepatoprotective effects of pecan nut shells on ethanol-induced liver damage. Exp Toxicol Pathol 2013;65(1–2):165–71. 127. Kumar Rajagopal S, Manickam P, Periyasamy V, Namasivayam N. Activity of Cassia auriculata leaf extract in rats with alcoholic liver injury. J Nutr Biochem 2003;14(8):452–8. 128. Kanuri G, Weber S, Volynets V, Spruss A, Bischoff SC, Bergheim I. Cinnamon extract protects against acute alcohol-induced liver steatosis in mice. J Nutr 2009;139(3):482–7. 129. Sun F, Xie ML, Zhu LJ, Xue J, Gu ZL. Inhibitory effect of osthole on alcohol-induced fatty liver in mice. Dig Liver Dis 2009;41(2):127–33. 130. Zhang J, Xue J, Wang H, Zhang Y, Xie M. Osthole improves alcohol-induced fatty liver in mice by reduction of hepatic oxidative stress. Phytother Res 2011;25(5):638–43. 131. Nanji AA, Jokelainen K, Tipoe GL, Rahemtulla A, Thomas P, Dannenberg AJ. Curcumin prevents alcohol-induced liver disease in rats by inhibiting the expression of NF-kappa B-dependent genes. Am J Physiol Gastrointest Liver Physiol 2003;284(2):G321–7. 132. Samuhasaneeto S, Thong-Ngam D, Kulaputana O, Suyasunanont D, Klaikeaw N. Curcumin decreased oxidative stress, inhibited NF-kappaB activation, and improved liver pathology in ethanol-induced liver injury in rats. J Biomed Biotechnol 2009;2009:981963. 133. Bao W, Li K, Rong S, Yao P, Hao L, Ying C, et al. Curcumin alleviates ethanol-induced hepatocytes oxidative damage involving heme oxygenase-1 induction. J Ethnopharmacol 2010;128(2):549–53. 134. McKim SE, Konno A, Gabele E, Uesugi T, Froh M, Sies H, et al. Cocoa extract protects against early alcohol-induced liver injury in the rat. Arch Biochem Biophys 2002;406(1):40–6. 135. Zhang F, Zhang J, Li Y. Corn oligopeptides protect against early alcoholic liver injury in rats. Food Chem Toxicol 2012;50(6):2149–54. 136. Kang MC, Ahn G, Yang X, Kim KN, Kang SM, Lee SH, et al. Hepatoprotective effects of dieckol-rich phlorotannins from Ecklonia cava, a brown seaweed, against ethanol induced liver damage in BALB/c mice. Food Chem Toxicol 2012;50(6):1986–91. 137. Lian LH, Wu YL, Song SZ, Wan Y, Xie WX, Li X, et al. Gentiana manshurica Kitagawa reverses acute alcohol-induced liver steatosis through blocking sterol regulatory element-binding protein-1 maturation. J Agric Food Chem 2010;58(24):13013–9. 138. Yuan G, Gong Z, Li J, Li X. Ginkgo biloba extract protects against alcohol-induced liver injury in rats. Phytother Res 2007;21(3):234–8. 139. Yao P, Li K, Song F, Zhou S, Sun X, Zhang X, et al. Heme oxygenase-1 upregulated by Ginkgo biloba extract: potential protection against ethanolinduced oxidative liver damage. Food Chem Toxicol 2007;45(8):1333–42. 140. Wang FS, Fan JG, Zhang Z, Gao B, Wang HY. The global burden of liver disease: the major impact of China. Hepatology 2014;60(6):2099–108. 141. Hong M, Kim SW, Han SH, Kim DJ, Suk KT, Kim YS, et al. Probiotics (Lactobacillus rhamnosus R0011 and acidophilus R0052) reduce the expression of toll-like receptor 4 in mice with alcoholic liver disease. PLoS One 2015;10(2):e0117451. 142. Bharrhan S, Koul A, Chopra K, Rishi P. Catechin suppresses an array of signalling molecules and modulates alcohol-induced endotoxin mediated liver injury in a rat model. PLoS One 2011;6(6):e20635. 143. Chen KH, Li PC, Lin WH, Chien CT, Low BH. Depression by a green tea extract of alcohol-induced oxidative stress and lipogenesis in rat liver. Biosci Biotechnol Biochem 2011;75(9):1668–76. 144. Kaviarasan S, Sundarapandiyan R, Anuradha CV. Epigallocatechin gallate, a green tea phytochemical, attenuates alcohol-induced hepatic protein and lipid damage. Toxicol Mech Methods 2008;18(8):645–52. 145. Yuan GGZ, Zhou X, Zhangq P, Sun X, Li X. Epigallocatechin-3-Gallate ameliorates alcohol-induced liver injury in rats. Int J Mol Sci 2006;7(7):204–19. 146. Yun JW, Kim YK, Lee BS, Kim CW, Hyun JS, Baik JH, et al. Effect of dietary epigallocatechin-3-gallate on cytochrome P450 2E1-dependent alcoholic liver damage: enhancement of fatty acid oxidation. Biosci Biotechnol Biochem 2007;71(12):2999–3006. 147. Li G, Ye Y, Kang J, Yao X, Zhang Y, Jiang W, et al. L-theanine prevents alcoholic liver injury through enhancing the antioxidant capability of hepatocytes. Food Chem Toxicol 2012;50(2):363–72. 148. Du J, He D, Sun LN, Han T, Zhang H, Qin LP, et al. Semen Hoveniae extract protects against acute alcohol-induced liver injury in mice. Pharm Biol 2010;48(8):953–8. 149. Yoo JH, Oidovsambuu S, Kim SM, Jeon NR, Yun JH, Kang K, Jho EH, Lee SB, Nho CW. Hepatoprotective effect of Handaeri-gomchi (Ligularia fischeri var. spiciformis Nakai) extract against chronic alcohol-induced liver damage in rats. Food Sci Biotechnol 2009;20(6):1655–61. 150. Yin HQ, Je YT, Kim YC, Shin YK, Sung S, Lee K, et al. Magnolia officinalis reverses alcoholic fatty liver by inhibiting the maturation of sterol regulatory element-binding protein-1c. J Pharmacol Sci 2009;109(4):486–95.
30 SECTION | I Overview of Liver Health
151. Yin HQ, Kim YC, Chung YS, Kim YC, Shin YK, Lee BH. Honokiol reverses alcoholic fatty liver by inhibiting the maturation of sterol regulatory element binding protein-1c and the expression of its downstream lipogenesis genes. Toxicol Appl Pharmacol 2009;236(1):124–30. 152. Khan AQ, Nafees S, Sultana S. Perillyl alcohol protects against ethanol induced acute liver injury in Wistar rats by inhibiting oxidative stress, NFkappa-B activation and proinflammatory cytokine production. Toxicology 2011;279(1–3):108–14. 153. Kim HK, Kim DS, Cho HY. Protective effects of Platycodi radix on alcohol-induced fatty liver. Biosci Biotechnol Biochem 2007;71(6):1550–2. 154. Zhang R, Hu Y, Yuan J, Wu D. Effects of Puerariae radix extract on the increasing intestinal permeability in rat with alcohol-induced liver injury. J Ethnopharmacol 2009;126(2):207–14. 155. Xiong Y, Yang Y, Yang J, Chai H, Li Y, Yang J, et al. Tectoridin, an isoflavone glycoside from the flower of Pueraria lobata, prevents acute ethanolinduced liver steatosis in mice. Toxicology 2010;276(1):64–72. 156. Zhao M, Du YQ, Yuan L, Wang NN. Protective effect of puerarin on acute alcoholic liver injury. Am J Chin Med 2010;38(2):241–9. 157. Ajmo JM, Liang X, Rogers CQ, Pennock B, You M. Resveratrol alleviates alcoholic fatty liver in mice. Am J Physiol Gastrointest Liver Physiol 2008;295(4):G833–42. 158. Kaviarasan S, Sundarapandiyan R, Anuradha CV. Protective action of fenugreek (Trigonella foenum graecum) seed polyphenols against alcoholinduced protein and lipid damage in rat liver. Cell Biol Toxicol 2008;24(5):391–400. 159. Dahiru D, Obidoa O. Pretreatment of albino rats with aqueous leaf extract of Ziziphus mauritiana protects against alcohol-induced liver damage. Trop J Pharm Res 2007;6(2):705–10. 160. Lieber CS, Leo MA, Cao Q, Ren C, DeCarli LM. Silymarin retards the progression of alcohol-induced hepatic fibrosis in baboons. J Clin Gastroenterol 2003;37(4):336–9. 161. Zhang W, Hong R, Tian T. Silymarin’s protective effects and possible mechanisms on alcoholic fatty liver for rats. Biomol Ther (Seoul) 2013;21(4):264–9. 162. Soylu AR, Altaner S, Aydodu N, Basaran UN, Tarcin O, Gedik N, et al. Effects of vitamins E and C supplementation on hepatic glutathione peroxidase activity and tissue injury associated with ethanol ingestion in malnourished rats. Curr Ther Res Clin Exp 2006;67(2):118–37. 163. Wang J, Guo J, Liu S. [Inhibitory effect of glycyrrhizin on NF-kappa B binding activity in CCl4 plus ethanol induced liver cirrhosis in rats]. Zhonghua Gan Zang Bing Za Zhi 1999;7(1):42–3. 164. Zhou T, Zhang YJ, Xu DP, Wang F, Zhou Y, Zheng J, et al. Protective effects of lemon juice on alcohol-induced liver injury in mice. Biomed Res Int 2017;2017:7463571. 165. Cheng N, Du B, Wang Y, Gao H, Cao W, Zheng J, et al. Antioxidant properties of jujube honey and its protective effects against chronic alcoholinduced liver damage in mice. Food Funct 2014;5(5):900–8. 166. Wang L, Wang BE, Wang J, Xiao PG, Tan XH. Herbal compound 861 regulates mRNA expression of collagen synthesis- and degradation-related genes in human hepatic stellate cells. World J Gastroenterol 2008;14(11):1790–4. 167. You H, Wang B, Wang T. [Proliferation and apoptosis of hepatic stellate cells and effects of compound 861 on liver fibrosis]. Zhonghua Gan Zang Bing Za Zhi 2000;8(2):78–80. 168. Yin C, Ma H, Wang A, Ma X, Jia J, Wang B. [Effect of compound 861 on tissue inhibitor of metalloprotenase 1 gene expression of HSC-T6 cells]. Zhonghua Gan Zang Bing Za Zhi 2002;10(3):197–9. 169. Baoen WTW, Jidong J, Hong M, Zhongping D, Xinmin L, Jia L, Aimin W, Linxue Q. Experimental and clinical study on inhibition and reversion of liver fibrosis with integrated Chinese and western medicine. CJIM 1999;5(1):6–11. 170. Yin SS, Wang BE, Wang TL, Jia JD, Qian LX. [The effect of Cpd 861 on chronic hepatitis B related fibrosis and early cirrhosis: a randomized, double blind, placebo controlled clinical trial]. Zhonghua Gan Zang Bing Za Zhi 2004;12(8):467–70. 171. Shimizu I, Ma YR, Mizobuchi Y, Liu F, Miura T, Nakai Y, et al. Effects of Sho-saiko-to, a Japanese herbal medicine, on hepatic fibrosis in rats. Hepatology 1999;29(1):149–60. 172. Cohen MR. Herbal and complementary and alternative medicine therapies for liver disease. A focus on Chinese traditional medicine in hepatitis C virus. Clin Liver Dis 2001;5(2):461–78. vii. 173. Dhiman RK, Chawla YK. Herbal medicines for liver diseases. Dig Dis Sci 2005;50(10):1807–12. 174. Schuppan D, Jia JD, Brinkhaus B, Hahn EG. Herbal products for liver diseases: a therapeutic challenge for the new millennium. Hepatology 1999;30(4):1099–104. 175. Mitra SK, Varma SR, Godavarthi A, Nandakumar KS. Liv.52 regulates ethanol induced PPARgamma and TNF alpha expression in HepG2 cells. Mol Cell Biochem 2008;315(1–2):9–15. 176. Mao YM, Zeng MD, Chen Y, Chen CW, Fu QC, Cai X, et al. [Magnesium isoglycyrrhizinate in the treatment of chronic liver diseases: a randomized, double-blind, multi-doses, active drug controlled, multi-center study]. Zhonghua Gan Zang Bing Za Zhi 2009;17(11):847–51. 177. Xiao ZW, Zhang W, Ma L, Qiu ZW. Therapeutic effect of magnesium isoglycyrrhizinate in rats on lung injury induced by paraquat poisoning. Eur Rev Med Pharmacol Sci 2014;18(3):311–20. 178. Testino G, Leone S, Ansaldi F, Borro P. Silymarin and S-adenosyl-l-methionine (SAMe): two promising pharmacological agents in case of chronic alcoholic hepathopathy. A review and a point of view. Minerva Gastroenterol Dietol 2013;59(4):341–56. 179. Stickel F, Patsenker E, Schuppan D. Herbal hepatotoxicity. J Hepatol 2005;43(5):901–10. 180. Nakagawa A, Yamaguchi T, Takao T, Amano H. [Five cases of drug-induced pneumonitis due to Sho-saiko-to or interferon-alpha or both]. Nihon Kyobu Shikkan Gakkai Zasshi 1995;33(12):1361–6. 181. DiCenzo R, Shelton M, Jordan K, Koval C, Forrest A, Reichman R, et al. Coadministration of milk thistle and indinavir in healthy subjects. Pharmacotherapy 2003;23(7):866–70.